Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
LITHUANIAN UNIVERSITY OF HEALTH SCIENCES MEDICAL ACADEMY Paulina Vaitkienė A STUDY OF TUMOR SUPPRESSOR GENE EXPRESSION AND PROMOTER METHYLATION FOR THE IDENTIFICATION OF PROGNOSTIC MARKERS IN GLIOBLASTOMA Doctoral Dissertation Biomedical Sciences, Biology (01B) Kaunas, 2013 Dissertation has been prepared at the Medical Academy of Lithuanian University of Health Sciences during the period of 2005–2013. Scientific Supervisor Prof. Dr. Dainius H. Pauža (Lithuanian University of Health Sciences, Medical Academy, Biomedical Sciences, Biology – 01B) Consultant Prof. Dr. Habil. Arimantas Tamašauskas (Lithuanian University of Health Sciences, Medical Academy, Biomedical Sciences, Medicine – 06B) 2 LIETUVOS SVEIKATOS MOKSLŲ UNIVERSITETAS MEDICINOS AKADEMIJA Paulina Vaitkienė GLIOBLASTOMŲ PROGNOZINIŲ ŽYMENŲ PAIEŠKA TIRIANT NAVIKĄ SLOPINANČIŲ GENŲ RAIŠKĄ BEI PROMOTORIŲ METILINIMĄ Daktaro disertacija Biomedicinos mokslai, biologija (01B) Kaunas, 2013 3 Disertacija rengta 2005–2013 metais Lietuvos sveikatos mokslų universiteto Medicinos akademijoje. Mokslinis vadovas prof. dr. Dainius H. Pauža (Lietuvos sveikatos mokslų universitetas, Medicinos akademija, biomedicinos mokslai, biologija – 01B) Konsultantas prof. habil. dr. Arimantas Tamašauskas (Lietuvos sveikatos mokslų universitetas, Medicinos akademija, biomedicinos mokslai, medicina – 06B) 4 CONTENTS ABBREVIATIONS ....................................................................................... 7 INTRODUCTION ......................................................................................... 9 1. 2. 3. AIM AND OBJECTIVES OF THE STUDY ................................... 10 ORIGINALITY OF THE STUDY ................................................... 11 REVIEW OF LITERATURE ........................................................... 13 3.1. Glioblastoma epidemiology and morphology ............................... 13 3.2. The concept of DNA methylation.................................................. 17 3.3. DNA methylation in gliomas ......................................................... 21 3.4. DNA methylation in medical research and practice ...................... 23 3.5. Genes selected for more detailed study ......................................... 25 4. MATERIALS AND METHODS ...................................................... 39 4.1. Patient tumor samples and characteristics ..................................... 39 4.2. RNA extraction .............................................................................. 39 4.3. DNA extraction .............................................................................. 41 4.5. cDNA synthesis ............................................................................. 44 4.6. DNA bisulphite modification ........................................................ 46 4.7. Sodium bisulphite sequencing ....................................................... 49 4.8. Methylation-specific PCR ............................................................. 53 4.9. Statistical analysis .......................................................................... 57 5. RESULTS ............................................................................................. 58 5.1. COX7A1, KRT81, AREG, NPTX2, and SPINT1 expression .......... 58 5.2. Bisulfite sequencing of genes promoters ....................................... 60 5.2.1. NTPX2 bisulfite sequencing .................................................... 60 5.2.2. AREG bisulfite sequencing ..................................................... 64 5.2.3. SPINT1 bisulphite sequencing ................................................ 65 5.2.4. COX7A1 bisulfite sequencing ................................................. 67 5.2.5. KRT81 bisulphite sequencing.................................................. 69 5.4. Characteristics of the study population tested for methylation of 11 genes …………………………………………………………………72 5.5. Gene methylation and its association with clinical variables and survival ................................................................................................. 74 5 5.5.1. AREG methylation .................................................................. 74 5.5.2. CASP8 methylation ................................................................. 76 5.5.3. CD81 methylation ................................................................... 78 5.5.4. DcR1 methylation.................................................................... 80 5.5.5. DR4 methylation ..................................................................... 82 5.5.6. GATA4 methylation ................................................................. 84 5.5.7. GATA6 methylation ................................................................. 86 5.5.8. hMLH1 methylation ................................................................ 88 5.5.9. NTPX2 methylation ................................................................. 89 5.5.10. TES methylation .................................................................... 91 5.5.11. TFPI2 methylation ................................................................ 93 5.6. Gene comethylation and its associations with survival ................. 96 DISCUSSION ............................................................................................ 104 CONCLUSIONS ........................................................................................ 113 REFERENCES ........................................................................................... 114 LIST OF PUBLICATIONS ....................................................................... 129 ACKNOWLEDGMENTS.......................................................................... 134 APPENDIX ................................................................................................ 135 6 ABBREVIATIONS AREG CASP8 CD81 CNS COX7A1 CpG ECM EGFR GBM GATA4 GATA6 G-CIMP H3K4 HIF-1 hMLH1 IDH KRT81 MBD MeCP2 amphiregulin caspase 8 CD81 molecule, TAPA-1 central nervous system cytochrome c oxidase subunit VIIa polypeptide 1 "p" phosphodiester bond between the cytosine and guanine nucleotides. cancer stem cells tumor necrosis factor receptor superfamily, member 10c deoxyribonucleic acid DNA methyltransferases tumor necrosis factor receptor superfamily, member 10a extracellular matrix epidermal growth factor receptor glioblastoma GATA binding protein 4 GATA binding protein 6 glioma- CpG island methylator phenotype lysine 4 on histone H3 hypoxic inducible factor 1 MutL homolog 1 isocitrate dehydrogenase enzyme keratin 81 DNA methyl-binding domain methyl CpG-binding proteins MGMT MMP-2 MMR MSP NPTX2 O6-methylguanine DNA methyltransferase matrix metalloproteinase 2 DNA mismatch repair system methylation-specific polymerase chain reaction neuronal pentraxin II CSC DcR1 DNA DNMT DR4 7 PCR PDGF RT-PCR SPINT1 TES TFPI-2 TRAIL VEGF VEGFR WHO 5hmC 5mC polymerase chain reaction platelet-derived growth factor reverse-transcription polymerase chain reaction serine peptidase inhibitor testin tissue factor pathway inhibitor 2 tumor necrosis factor-related apoptosis-inducing ligand vascular endothelial growth factor vascular endothelial growth factor receptor World Health Organization 5-hydroxymethylcytosine 5-methylcytosine 8 INTRODUCTION Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Patients with glioblastoma have a median survival time of only 12-15 months (1, 2), and prognosis remains extremely poor despite multimodal treatment by surgery, radiotherapy and, chemotherapy. Generally, prognostically favorable clinical factors, such as young age and a good Karnofsky performance score at diagnosis are associated with a better prognosis. However, various clinical characteristics fail to explain a variation in disease progression and response to treatment in histologically diagnosed glioblastomas. There is increasing interest in identifying the molecular markers that better capture the status of a tumor in order to improve the existing predictions (3). The identification of methylated genes in cancer may provide an insight in the molecular mechanisms of tumor development and might reveal new tools to define the markers of prognostic significance (4). For example, the epigenetic silencing of the tumor suppressor gene O6-methylguanine DNA methyltransferase (MGMT), which encodes a DNA repair enzyme, sensitizes cancer cells to alkylating agents and is associated with a significantly improved outcome in GBM patients treated with radiotherapy and temozolomide (5). Recently, several microarray-based GBM studies have identified the gene CpG rich promoter-associated sequences that are frequently methylated. When hypermethylation occurs within the promoter region of a gene, it could inhibit its expression, contributing to the inactivation of tumor suppressor genes in cancer and provide the cell with a growth advantage (6). The list of genes reported as being silenced in gliomas by aberrant promoter methylation is growing and includes targets involved in glioma development and progression. Up to now, a lot of genes were identified to be differentially methylated in a subset of gliomas, enabling the discrimination of tumors with different biological and clinical properties according to the methylation pattern (2, 7, 8). However, studies are usually limited because of a small sample of glioblastomas and scarce clinical data. In order to identify the molecular markers relevant to the development, diagnosis, and prognosis of glioblastoma, the analysis of 14 genes (AREG, CASP8, CD81, COX7A1, DcR1, DR4, GATA4, GATA6, hMLH1, KRT81, NPTX2, SPINT1, TES, and TFPI-2) was performed. The products of these genes inhibit cellular migration and metastasis, regulate cell proliferation, and protect the genome from mutagenesis. The main objective of our study was to further characterize the role of the latter genes in the pathogenesis of glioblastoma and to evaluate its clinical importance. 9 1. AIM AND OBJECTIVES OF THE STUDY The aim of this study was to determine aberrant promoter methylation of tumor suppressor genes in tumor tissue DNA from patients with glioblastoma multiforme and to evaluate the associations between the methylation profile of genes, patients’ clinical characteristics, and prognostic value. The objectives of the study were as follows: 1. To investigate the expression of 5 genes (AREG, COX7A1, KRT81, NPTX2, and SPINT1) in 22 different glioblastoma samples and to compare with the expression of these genes in control brain tissue samples; 2. To determine if the differences in the expression of AREG, COX7A1, KRT81, NPTX2, and SPINT1 genes are associated with the promoter methylation of these genes; 3. To determine the methylation frequency of 11 genes (AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, TES, and TFPI2), involved in cell motility, growth, adhesion, apoptosis, and repair processes, in 100 glioblastoma tissue samples; 4. To evaluate the associations between AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, TES, and TFPI2 gene promoter methylation and clinical characteristics of patients with glioblastoma. 5. To determine the relationship between AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, TES, and TFPI2 gene promoter methylation and patients’ survival after surgery; 6. To identify epigenetic markers and their combinations that could be significant in the prognosis of patients with glioblastoma. 10 2. ORIGINALITY OF THE STUDY Glioblastoma is one of the most aggressive tumors with a poor prognosis. The epigenetic events that regulate gene expression have clearly emerged as a fundamental mechanism in developmental biology and in the pathogenesis of cancer. For example, multiple genes that affect numerous cellular pathways are silenced by hypermethylation in cancer, and studies of these genes have increased our understanding of how cancer develops and progresses. In 2007, Mueller et al. succeeded in unveiling novel candidate genes, which are epigenetically regulated in glioblastomas, by using the pharmacologic manipulation of glioma cells with the demethylating agent 5´-aza-dC combined with genome wide expression profiling (7). To confirm the importance of these genes in gliomagenesis, in this study, a more detailed analysis of COX7A1, SPINT1, AREG, NPTX2, and KRT81 gene expression and their promoter methylation in glioblastoma samples was performed for the first time. Genes were evaluated by using the following methods: reverse-transcription polymerase chain reaction (RT-PCR), bisulfite sequencing, and methylation-specific PCR (MSP). Potential epigenetic markers in glioblastoma – AREG and NPTX2 – the expression of which was found to be associated with promoter methylation have been identified. Moreover, in this study, the methylation frequency of 11 genes (AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, TES, and TFPI2), involved in cell motility, growth, adhesion, apoptosis, and repair processes, was determined in 100 glioblastoma tissue samples. The data on the frequency of methylation of these genes in glioblastoma are scarce to evaluate the usability and suitability of new epigenetic markers in molecular diagnosis of these tumors and prognosis of the disease. To our knowledge, the methylation frequencies of the AREG, DcR1, GATA4, NPTX2, and TFPI2 gene promoters in glioblastoma have not been reported in literature; therefore, we believe that our study was the first to determine the methylation frequencies and significantly contributed to the existing knowledge on epigenetic markers in glioblastoma and the process of gliomagenesis. Our study differs from other published studies by a sample size, analysis of clinical data, and identification of new methylation markers and their combinations that could be used in the diagnosis and prognosis of glioblastoma. 11 To our knowledge, this study is the first to report the significant associations between the methylation of the AREG, CASP8, GATA6, and TFPI2 gene promoters and patients’ survival after surgery. After a detailed investigation of these genes and confirmation of the data in larger scale studies, they could be used for the molecular typing of glioblastomas and more accurate diagnosis to tailor the treatment strategies and prognosticate the course of the disease. Moreover, this study is one of the first studies where not only the impact of the methylation of single gene promoters on survival has been studied, but also associations between the comethylation of several genes and patients’ clinical characteristics have been determined. With the help of comprehensive statistical analysis, a unique set of 6 genes (AREG, CASP8, DR4, GATA4, GATA6, and TFPI2) has been identified; the methylation studies of the promoters of this gene combination would help reliably predict survival as compared with the analysis of individual genes. The importance of this combination of genes in the diagnosis and prognosis of disease course in glioblastoma needs to be evaluated and validated in more detailed studies with a larger cohort of patients with glioblastoma. In this study, the fundamental investigations were linked to clinical data. The study conclusions are undoubtedly useful for clinical practice, and the obtained results provide additional information for the future planning of new clinical trials. 12 3. REVIEW OF LITERATURE 3.1. Glioblastoma epidemiology and morphology Central nervous system (CNS) tumors in Europe have an incidence of approximately 6-7 per 100 000 persons per year and account for 2% of all cancer-related deaths (9). The diagnosis and grading of primary brain tumors follows the World Health Organization (WHO) classification (10). The WHO classification indicates malignant potential, response to treatment, and survival. Gliomas are tumors that are thought to arise from glial progenitor and glial cells. These tumors account for 32% of all primary brain tumors and, and 80% of all malignant primary brain tumors diagnosed in the United States (11). The overall annual incidence of gliomas varies from 5 to 6 cases per 100 000 worldwide (12). Glial tumors are categorized into 4 grades. Grade I and II low-grade astrocytomas are slow-growing, less aggressive tumors, while grade III and IV high-grade gliomas are malignant tumors, characterized by a high proliferation rate (grade III) and the presence of necrotic tissue and/or angiogenic activity (grade IV) (13). Glioblastoma is also known as grade IV astrocytoma or glioblastoma multiforme. The term multiforme refers to the vast intratumoral heterogeneity seen in the disease. The histopathology of glioblastoma shows brisk mitotic activity, microvascular proliferations, thrombosis, and necrosis (14). Glioblastoma multiforme accounts for up to 60% of all malignant primary brain tumors in adults, occurring in 2-3 cases per 100 000 in Europe and North America (15). It is one of the most devastating and lethal forms of human cancer. GBM presents some of the greatest challenges in the management of cancer patients worldwide, despite notable recent achievements in oncology. Even with aggressive surgical resections using state-of-the-art preoperative and intraoperative neuroimaging, along with recent advances in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal: survival time after diagnosis is about 1 year (16). Only 3%–5% of patients survive longer than 5 years (17). Nevertheless, residual tumor causes relapse in almost all cases. GBM most frequently recurs after a median survival time of 32 to 36 weeks (18). Although GBM can manifest itself at any age, it preferentially occurs in adults, with a peak age of incidence between 45 and 70 years (19). This type of cancer is more common in men than women. It most frequently involves the brain hemispheres, but it can also affect basal ganglia and the brain stem (19). The etiology of malignant glioma is unknown. Therapeutic ionizing irradiation has been confirmed as a sole unequivocal risk factor for the development of 13 glioblastoma. No association between exposure to electromagnetic fields and development of brain tumors has been established so far. Further, no definite association between viral infection or diet (e.g., intake of N-nitroso compounds) and malignant glioma has been proven as well (14). Invasion An additional important feature of malignant glioma include its great tendency to infiltrate into normal brain tissue, rendering the condition incurable by surgery alone (19). Migrating glioma cells are relatively resistant to cytotoxic therapy (chemotherapy, irradiation) because the processes of metabolism are reduced as compared to nonmigrating tumor cells. The invasion of glioma cells requires interaction with the extracellular matrix (ECM) and with surrounding cells of the healthy brain tissue. The brain ECM is mainly composed of soft components such as glycosaminoglycans, proteoglycans, tenascin-C, and thrombospondin (14). Rigid components like collagen, fibronectin, or laminin are limited to perivascular and vascular areas in the brain. Migrating glioma cells may degrade the soft components of ECM by proteases, namely the matrix metalloproteinases, serine proteases, and cysteine proteases (14). The outer GBM border usually observed in T1-weighted gadolinium magnetic resonance imaging does not delineate the true dimension of the tumor. Moreover, tumor cells diffusely invade the normal brain and may be detected beyond that border (19). Hypoxia and angiogenesis Vascular proliferation and tissue necrosis are characteristic features of malignant gliomas, particularly of glioblastoma. These features are most likely the consequence of rapidly increasing tumor mass that is inadequately oxygenized by the preexisting vasculature (14). Hypoxia-related genes are significantly associated with necrosis in human glioblastoma specimens, which suggests that there is a possible link between glioblastoma necrosis and hypoxia. Hypoxic cells are known to overexpress hypoxic inducible factor 1 (HIF-1), vascular endothelial growth factor (VEGF), and matrix metalloproteinase 2 (MMP-2) (20). HIF-1 is translocated to the nucleus and induces the transcription of target genes involved in angiogenesis, migration, proliferation, and cell survival. The downstream effectors of HIF-1 include VEGF and vascular endothelial growth factor receptor (VEGFR). VEGF is a major angiogenic growth factor. It binds to and activates the tyrosine kinase receptors of the VEGFR family. Activated 14 tyrosine kinase activates downstream pathways that induce proliferation and migration of vascular cells (14). Considering the morphology of glioblastoma necrosis, these factors could cause tumor cell migration to form pseudopalisades and microvascular hyperplasia in perinecrotic areas, which are associated with poor prognosis in cancer (20). Tumor cells upregulate hypoxia-associated factors that induce angiogenesis. In malignant gliomas, newly formed vessels do not show a normal phenotype, but appear as bizarre vascular proliferates. It is assumed that these bizarre vascular proliferates are not suitable to sufficiently oxygenize the tumor tissue, resulting in a vicious circle of hypoxia and aberrant angiogenesis (14). Glioblastoma cancer stem cell hypothesis Traditionally, it was thought that the adult human brain does not contain precursor cells, and it was assumed that brain tumors derive from mature parenchymal cells (14). An increasing body of evidence has strongly supported the concept that a subpopulation of cancer cells in each tumor has a greater potential of cancer initiation and repopulation. These cells are known as cancer stem cells (CSCs) or tumor-initiating or propagating cells because they share some critical characteristics with normal stem cells (21). CSCs are defined as tumor cells with stem cell properties, i.e., asymmetric cell division, infinite growth, and multipotency, that later differentiate into rapidly proliferating progenitor-like and more differentiated cells, defining the histopathology of the tumor (22). The CSC hypothesis also suggests that resistance to chemo- and radiotherapy is driven by CSCs and thus challenges the present diagnostic and therapeutic strategies (22). It has been suggested that cancer stem cells are the main proliferating cell population in malignant gliomas responsible for tumor growth and progression (14). GBM stem cells may be resulted from genetic and epigenetic changes in neural stem/progenitor cells or the differentiated cells such as astrocytes after a series of mutations or epigenetic reprogram. Although the origin of GBM stem cells is not clearly defined, they share similar properties with normal neural stem cells that endow these cells with key traits in carcinogenesis (21). Pathogenesis of glioblastomas Gliomas, like all human tumors, arise by a step-wise accumulation of genetic events that inactivate tumor suppressor genes and activate oncogenes (23). Glioblastoma may develop from diffuse low-grade or anaplastic astrocytomas (secondary glioblastoma), but more frequently, they 15 manifest de novo, without a less malignant precursor lesion (primary glioblastoma) (14). Primary and secondary GBMs are histologically indistinguishable, but at the molecular level, each type demonstrates distinct patterns of genetic alterations and these have long been thought to represent different routes of molecular pathogenesis to a common histopathologic end point (24). Two genetic alterations have been detected to occur at a significant rate in primary GBM: amplification or activating mutation of epidermal growth factor receptor (EGFR) and loss of chromosome 10 (23). The activation of growth factor receptors – EGFR, alpha-type plateletderived growth factor receptor, or MET – by mutation and amplification inhibits the functions of tumor suppressors such as p53 and Rb (24). The tumor suppressor gene PTEN (phosphatase and tensin homolog) is mutated in the 10q23 region in approximately one-third of primary GBMs (19). Mutations in this gene have been described only in malignant gliomas and are rarely associated with p53 mutations (19). Other frequent mutations in primary GBM affect the CDK cell-cycle-regulator genes CDK4/6 and CDKN2A/CDKN2B (19). In summary, primary glioblastoma typically arises with a complement of mutations that, one way or another, serves to activate progrowth pathways and inactivate or suppress tumor suppressors p53 and Rb. Because primary GBMs arise de novo, there is little information on the sequence of events during pathogenesis (24). Other GBMs arise by the step-wise progression through a series of lower grade lesions (low-grade astrocytoma, anaplastic astrocytoma) and are called secondary GBMs (23). A number of alteration have been noted in lower grade lesions, including the loss of the tumor suppressors p53, p15, p16, and Rb and overexpression of the platelet-derived growth factor (PDGF) (23). Somatic mutations of the isocitrate dehydrogenase enzymes (IDH1 and IDH2) appears to play a critical and early role of these gliomas (24). The recent finding of mutations in IDH genes in 70%-80% of grade II and III astrocytomas, as well as some glioblastomas, has provided a unifying mechanism for early gliomagenesis. As IDH enzymes are typically associated with the Krebs cycle, they play a multitude of other roles in normal cellular biology, such as contributing to cellular defenses against oxidative damage and, potentially, regulating epigenetic modifiers (25). The incidence of IDH mutation in primary GBM is low (~5%), reinforcing the notation of a distinct pathogenesis associated with tumors arising in this clinical scenario (24). Despite the genetic differences, no differences in sensitivity to conventional chemotherapy between primary and secondary glioblastoma multiforme have been reported. The molecular and genetic aberrations in 16 There are not many molecular factors that contribute to long-term survival. Among the molecular markers, TP53 mutation, infrequent amplification of the EGFR gene, combined LOH 1p/19q, loss of chromosome arm 19q, and low rate of tumor proliferation have been associated with long-term survival in GBM (26, 27). MGMT promoter methylation is seen in ~50% of glioblastomas and has been linked to prolonged progression-free and overall survival (1). PTEN protein expression was a favorable factor for long-term survival (27). In addition, mutations in the IDH1 and IDH2 genes have recently been reported in a subset of glioblastomas from younger patients and linked to a more favorable survival outcome (29). So far, the molecular and cellular mechanisms of long-term survival in glioblastoma patients are not known. 3.2. The concept of DNA methylation Glioblastomas arise by a step-wise accumulation of the events that inactivate tumor suppressor genes and activate oncogenes. These can occur by both epigenetic regulatory mechanisms and structural genomic alterations such as overexpression and amplification activating oncogenes, and transcriptional silencing and genomic deletion inactivating tumor suppressor genes (23). Epigenetics has emerged as an important field in biomedical research in the last decade, describing the novel mechanisms of normal biological processes as well as disease pathogenesis. Epigenetics can be defined as the study of heritable changes of a phenotype such as the gene expression patterns of a specific cell type that are not caused by changes in the nucleotide sequence of genetic code itself (30). Epigenetic regulatory mechanisms are as follows: 1. Posttranslational histone modification; 2. ATP-dependent chromatin remodeling complexes; 3. Polycomb/trithorax protein complexes; 4. Small and other noncoding RNA (siRNA and miRNA); 5. DNA methylation. These mechanisms are discrete, but interact closely in order to regulate gene expression. Many of the changes, particularly those resulting from DNA methylation and histone deacetylation, affect gene expression and genome stability through the inappropriate regulation of a local chromatin structure. Furthermore, recent data suggest that epigenetic changes are involved in the earliest phases of tumorigenesis, and that they may predispose stem (progenitor) cells to subsequent genetic and epigenetic changes that are 17 involved in tumor promotion. Given the observed frequency of DNA methylation changes in tumorigenesis and the inherent stability of these molecular abnormalities, these events may provide ideal biomarkers for molecular diagnostics and early detection of cancer (19). DNA methylation is currently the most widely studied form of epigenetic programming. This postreplication modification is almost exclusively found on the 5 position of the pyrimidine ring of cytosines in the context of the dinucleotide sequence CpG (31) (Figure 3.2.1). Figure 3.2.1. Mechanism of CpG methylation (A) CpG islands consist of nucleotide bases where cytosine (C) residues are followed by guanine (G) in a genomic DNA sequence. (B) Methylation of CpG islands is catalyzed by DNA methyltransferases (DNMTs), which transfer the methyl group from S-adenosylmethionine (SAM-CH3) to cytosine. The methylation reaction yields S-adenosyl homocysteine (SAH) and 5-methylcytosine (5mC) (32). In the term “CpG sites,” “p” represents the phosphodiester bond between the cytosine and guanine nucleotides. The methylation of CpG rich clusters, called CpG islands, is important because they are found in the promoter region and first exons of many genes. CpG islands are regions of ~500 bp to 1 kbp where CpG nucleotides are about 5 times more abundant compared with the rest of the genome (33). Although CpG islands account for approximately 1% of the total human genome, they are present in >50% of 18 human gene promoters indicating their functional importance in transcriptional control. DNA methylation is mediated by 1 of 3 DNA methyltransferases (DNMTs), which catalyze the addition of a methyl group from a universal methyl donor, S-adenosyl-L-methionine, to the 5-carbon position of a cytosine (31). DNMT1 is considered to be primarily responsible for the maintenance of this epigenetic modification and copies the pre-existing methylation pattern onto the daughter strand after DNA replication. DNMT3A and DNMT3B are de novo methyltransferases and can also complete the methylation process and correct the errors (31). CpG islands are variable methylated as a mechanism to regulate gene transcription. They are mostly nonmethylated associated with the maintenance of an open chromatin structure and a potentially active state of transcription (30). Methylation above normal levels is referred to as hypermethylation. The hypermethylation of CpG islands in a gene promoter blocks the ability of transcription factors to interact with the promoter and inhibits gene expression (32) (Figure 3.2.2.). Several mechanisms have been proposed to be associated with transcriptional repression by DNA methylation. The first mechanism involves direct interference with the binding of specific transcription factors to their recognition sites in their respective promoters. Several transcription factors, including AP-2, c-Myc/Myn, the cyclic AMP-dependent activator CREB, E2F, and NFκB, recognize sequences that contain CpG residues, and binding of each has been shown to be inhibited by methylation (35). The second mode of repression involves a direct binding of specific transcriptional repressors to methylated DNA (35). In addition to the proteins that create and maintain the methylation pattern, there are proteins that recognize methylated DNA and interact with it. DNA methyl-binding domain (MBD1-4) proteins or methyl CpG-binding proteins (MeCP2) recognize and bind to methylated DNA. They recruit transcriptional corepressors such as histone-deacetylating complexes, polycomb proteins, chromatin-remodeling complexes and attract chromodomain-binding proteins (30). Methylated DNA can also be bound by some zinc finger proteins such as Kaiso and more recently discovered ZBTB4 and ZBTB38 proteins that are also able to repress transcription in a methylation-dependent manner (30). 19 Figure 3.2.2. Epigenetic patterns in normal and cancer cells (A) DNA methylation. In normal cells, nearly all of the CpG dinucleotides are methylated whereas CpG islands, mostly residing in 5′ regulatory regions of genes, are unmethylated. In cancer cells, many CpG islands become hypermethylated, in conjunction with silencing of their cognate genes, while global hypomethylation, mostly at repetitive elements, occurs. (B) Chromatin and histone modification. Active genes are associated with acetylation of histone tails, methylation of lysine 4 on histone H3 (H3K4), and nucleosome depletion at their promoters. The promoters of silenced genes (drawn here in conjunction with DNA hypermethylation) become associated with nucleosomes, lose acetylation and H3K4 methylation marks, and gain repressive methylation marks such as lysine 9 or 27 on histone H3, which recruit repressive complexes. Methylated DNA binding proteins link methylated DNA with the histone modification and nucleosome remodeling machineries (not shown) (34). DNA methylation can also affect histone modifications and chromatin structure, which, in turn, can alter gene expression. DNA methylation and histone modifications function in a close interplay with nucleosome remodeling and positioning complexes that bind specifically modified histones and methyl CpG-binding proteins, and move nucleosomes on DNA by ATP-dependent mechanisms (34). Nonmethylated CpG island promoters are usually hypersensitive to nucleases and are relatively depleted of nucleosomes, whereas methylated promoters have nucleosomes on them and are nuclease resistant (34). 20 DNA methylation is possibly a reversible step, and there are indications that under some conditions, the methylated DNA pattern is a balance between active demethylation and remethylation. Demethylation in the absence of DNA replication normally occurs in the zygote and in the development of germ cells, but it also is active in malignant cells. The molecular mechanisms for demethylation remain unclear, though recent studies suggest that DNMT3A and 3B may be involved (31). In many cases, methylated and silenced genes can be reactivated using DNA methylation inhibitors such as 5-azacytidine. However, it should be noted that an unmethylated state of a CpG island does not necessarily correlate with the transcriptional activity of the gene, but rather that the gene can be potentially activated. On the other hand, the presence of methylation alone does not necessarily induce silencing of nearby genes. Only when a specific core region of the promoter that is often, but not necessarily, spanning the transcription start site becomes hypermethylated, the expression of the associated gene is modified (30). 3.3. DNA methylation in gliomas In glioblastomas, the changes in methylation patterns can be seen, which are characterized by a genome-wide loss of methylation combined with locus-specific hypermethylation. Hypomethylation may promote tumor growth by the activation of oncogenes, loss of imprinting, or promotion of genomic instability. CpG island promoter methylation occurs in genes with diverse functions related to tumorigenesis and tumor progression, including cell-cycle regulation, DNA repair, apoptosis, angiogenesis, invasion, and drug resistance. CpG hypermethylation in gliomas may also occur in genes that are not expressed in the brain, suggesting not all events of CpG island methylation are functionally important for tumorigenesis (33). Several epigenetic alterations have been described in glioblastoma. Among them, the methylation of MGMT has been the most studied. MGMT encodes a DNA repair protein that removes alkyl adducts at the O6 position of a guanine and protects normal cells from carcinogens. Unfortunately, it can also protect tumor cells from chemotherapeutic alkylating agents such as temozolomide. von Deimling et al. in their article summarized that GBM patients with a hypermethylated MGMT promoter demonstrated the survival rates of 49% and 14% at 2 and 5 years, respectively, when treated with concomitant and adjuvant temozolomide and radiotherapy. In contrast, the estimated 2- and 5-year survival rates were only 24% and 5%, respectively, 21 in similar patients that were initially treated with radiotherapy alone. GBM patients whose tumors lacked MGMT hypermethylation demonstrated the 2and 5-year survival rates of 15% and 8%, when they received combined radiochemotherapy, and the rates decreased to 2% and 0% if radiotherapy was administered alone (36). There are data that different-grade gliomas show differences in the methylation status of various genes (37-39). In this context, the methylation of the DNA repair gene MGMT is more frequently observed in secondary GBMs (about 75%) than primary GBMs (36%) or low-grade astrocytomas (48%) (19). Secondary GBMs have a higher overall frequency of promoter methylation compared to primary GBMs at least for the promoters of p14, p16, RB1, MGMT, and TIMP-3. Low-grade gliomas and secondary GBMs show PTEN promoter methylation (33). The progression of glioma over time is associated with distinct epigenetic patterns. Malignant progression and shorter survival in astrocytoma are associated with p14, but not MGMT hypermethylation, suggesting that these are 2 distinct pathways in astrocytoma with different clinical consequences (33). The association between methylation levels and WHO grading may directly suggest that hypermethylation reflects a potential of tumor aggressiveness. On the other hand, there are data that methylation profiles are remarkably stable across glioma evolution, even during anaplastic transformations, suggesting that epigenetic alterations occur early during gliomagenesis (6). There are some data that the analysis of the concomitant hypermethylation of several genes allows better prediction of survival than that of one gene (9). Noushmer et al. reported that gliomas could be stratified by the glioma CpG island methylator phenotype (G-CIMP) status into 2 distinct subgroups with different molecular and clinical phenotypes. These molecular classifications have implications for differential therapeutic strategies for glioma patients (8). Genes involved in invasion and metastasis can also be affected by promoter hypermethylation in gliomas. The CpG promoter hypermethylation of the protocadherin-gamma subfamily is frequently observed in astrocytomas, glioblastomas, and glioma cell lines (33). The significance of CpG island promoter hypermethylation in GBM pathogenesis is emphasized by the observation of epigenetically mediated inactivation of a wide variety of genes associated with tumor suppression (RB1, VHL, EMP3), cell cycle regulation (p16, p15), DNA repair (MGMT, hMLH1), tumor invasion, and apoptosis (19). 22 3.4. DNA methylation in medical research and practice With a growing body of knowledge about the pathogenesis of glioblastoma, there is hope for the development of novel therapeutic agents that may effectively treat patients. DNA methylation is potentially preventable or possibly reversible. For example, blocking DNA methylation by inhibiting DNMTs results in the demethylation of CpG islands in daughter cells, with subsequent restored expression of tumor suppressor genes and abrogation of tumor growth (40). Successful conventional chemotherapy depends on the activation of proapoptotic genes that respond to cytotoxic agents leading to cell death. DNA methylation of these proapoptotic genes can block cell death from occurring leading to chemotherapeutic resistance, thus the reactivation of epigenetically silenced apoptotic genes can increase the efficacy of chemotherapy (41). Many compounds have been discovered to target proteins that control DNA methylation. Some of them are already being used clinically. The most investigated DNA methylation inhibitors are as follows (41): 1. 5-Azacytidine (5-Aza-CR; Vidaza) is a nucleoside analog that incorporates into RNA and DNA and is FDA approved to treat high-risk myelodysplastic syndromes (MDS) patients; successful clinical results have recently been reported. 2. 5-Aza-2 deoxycytidine (5-Aza-CdR; Decitabine) is the deoxy derivative of 5-Aza-CR and is incorporated only into DNA. 3. Zebularine, a cytidine analog that acts similarly to 5-Aza-CR, but has lower toxicity, increased stability and specificity. 4. S110, a decitabine derivative that has increased stability and activity, which has shown promise in preclinical studies. Two clinically used compounds with DNA demethylating activities – azacitidine (5-azaCR, Vidaza; Celgene, Summit, NJ, USA) and decitabine (5-aza-2′-deoxycytidine, Dacogen; SuperGen, Dublin, CA, USA) – are structurally similar to cytosine nucleosides (42). These inhibitors are incorporated into DNA and trap the DNMT leading to a covalent proteinDNA adduct, depletion of DNMTs, and subsequent demethylation of genomic DNA during replication. Although proven to be potentially effective as antitumor agents in laboratory experiments and in clinical trials, both inhibitors are unstable in solution and can be toxic (43). DNA methylation inhibitors act during S-phase of the cell cycle, thus they preferentially affect rapidly growing cells. In addition to inhibiting DNA methyltransferase activity, azanucleosides also act through unspecific mechanisms, which likely contribute to their clinical effectiveness (41). 23 Administration of a combination of DNMT and HDAC could have a synergistic effect on gene activation and could allow lower doses of each drug to be used (33). Zebularine is a proven nucleoside DNMT inhibitor effective at reactivating the silenced genes in vitro and in vivo with a high specificity for cancer cells relative to normal cells. It was demonstrated that Zebularine selectively sensitizes the brain tumor cells that are deficient in DNAdependent protein kinase. The sensitization of cell killing by Zebularine is mediated by a combination of DNA repair and cell cycle checkpoint defects in DNA-dependent protein kinase-deficient glioblastoma cells (43). The methylation-induced silencing of cancer-testis antigens, such as NYESO-1, can protect cancer cells from being recognized by T cells. Treating cancer cells with demethylating agents can induce the expression of these antigens, allowing a reaction by engineered lymphocytes suggesting that epigenetic therapy can be combined with immunotherapy for better results (41). Currently available DNA methylation inhibitors lead to overall DNA methylation inhibition. Global hypomethylation may lead to the activation of oncogenes and/or increased genomic instability. Recently, DNA methylation inhibitors that targeted specific genes or the groups of genes are being developed. Another way is to identify and use demethylases. DNA methylation is involved in key cellular processes important to tissue-specific gene expression, cell differentiation, and development. DNA methylation patterns are frequently perturbed in human diseases such as imprinting disorders and cancer (44). It is largely unknown about the mechanisms that control methylation levels and prevent from DNA methylation at CpG islands. After fertilization and at certain points of the embryonic development, a large number of the methylation markers are erased (45). Importantly, despite an intense search, no direct DNA demethylase has been identified yet (44). Until recently, the only known epigenetic marker of DNA itself was 5mC, which is converted by DNA methyltransferases preferentially at CpG dinucleotides (46). 5Hydroxymethylcytosine (5hmC), which is considered to be the sixth base of the genome of higher organisms, has recently been identified (45). The recent discovery that the 3 members of the TET protein family can convert 5mC into 5hmC has provided a potential mechanism leading to DNA demethylation (44). 24 Figure 3.4.1. Demethylation model The canonical DNA nucleosides dG, dA, dT, and dC. Cytosine can be modified to mC and hmC in mammalian tissues. hmC could be further oxidized to the putative demethylation intermediates fC and caC, but have never been found in vivo (45). Despite a lot of promising epigenetic data, they have not been used in routine diagnostic practice yet. The hypermethylation of gene promoters may be considered the tip of iceberg of deregulation of complex epigenetic mechanisms that contribute to GBM onset (19). Whole genome, transcriptome, and methylome sequencing is currently at the horizon of clinical research and could be performed even in small-sized tumor tissue samples (47). Unraveling the epigenetic alterations opens up possibilities for discovering new biomarkers for cancer detection and prognosis (19). Restoring epigenetically altered pathways will probably lead to the development of new therapeutic tools in GBM. Blood plasma in cancer patients contains DNA derived from cells due to necrotic or apoptotic cancer cells releasing genomic DNA. This potentially provides a less invasive method for the detection of biomarkers. Aberrantly methylated cancer genes found in plasma could be one such type of biomarkers (33). 3.5. Genes selected for more detailed study The cytosine methylation of CpG dinucleotides in gene promoters is a common cause of DNA silencing and transcriptional repression that can 25 modulate the clinical characteristics of glioblastoma. The best known is MGMT promoter methylation determining tumor response to DNA alkylating agents and being an independent prognostic factor for patients’ survival (48). However, several widely described genes can be seen as only a partial picture of the methylation changes, and there may be many more genes that need to be clarified. Recent methylome studies of brain tumors have disclosed a list of new epigenetically modified genes associated with gliomagenesis and different glioma clusters (2, 6, 8). The latter studies illustrate a glioblastoma profile being constructed via methylation of a multiple set of genes, forming networks attributed to different pathogenesis pathways. It has been suggested that genes, such as AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, TES, and TFPI2 could be important in gliomagenesis. Table 3.5.1. Functions and reported methylation frequency of the selected genes in glioblastoma Gene Location Functions of encoded proteins Autocrine growth factor and mitogen for astrocytes, Schwann cells, and fibroblasts Responsible for the TNFRSF6/FAS 2q33 mediated and TNFRSF1A induced CASP8 apoptosis Encoded protein is a cell surface 11p15.5 glycoprotein that is known to form CD81 a complex with integrins May protect cells against TRAIL mediated apoptosis by competing 8p22 DcR1 with TRAIL-R1 and R2 for binding to the ligand Receptor for the cytotoxic ligand 8p21 DR4 TNFSF10/TRAIL 8p23.1 Transcriptional activator GATA4 18q11.1 Transcriptional activator GATA6 Modulates postreplicative DNA 3p21.3 hMLH1 mismatch repair system 7q21.3 Modulates long-term cell plasticity NPTX2 May play a role in cell adhesion, cell spreading and in the 7q31.2 TES reorganization of the actin cytoskeleton May play a role in the regulation of 7q22 plasmin-mediated matrix TFPI2 remodeling ND, no data on DNA methylation in glioblastomas. AREG 4q13 26 GBM methylation in %, (literature source) ND 10-30 (49, 50) 54 (2, 6) ND 25-70 (2, 26, 49) ND 30-48 (2, 50-52) 0-15 (53, 54) ND 58-69 (2, 7) ND AREG The AREG (amphiregulin, SDGF, schwannoma-derived growth factor) gene is located on chromosome 4q13.3. AREG is a member of the epidermal growth factor family. It is a bifunctional growth modulator: it interacts with the EGF/TGF receptor to promote the growth of keratinocytes and normal lung epithelial cells in the presence of extracellular matrix, but inhibits the growth of certain aggressive carcinoma cell lines, some transformed cell lines, and lung epithelial cells in the absence of extracellular matrix (55, 56). Thus, AREG can either stimulate or inhibit the growth of lung cancer cells, depending on the biological settings (56). Various studies have highlighted the functional role of AREG in several aspects of tumorigenesis, including self-sufficiency in generating growth signals, limitless replicative potential, tissue invasion and metastasis, angiogenesis, and resistance to apoptosis (57). On the other hand, there is an growing body of data about AREG promoter methylation in various types of tumors: gastric (58), high-grade serous carcinomas (59), and human bladder tumor cell line (T24) (60). AREG is differentially methylated in low-grade gliomas (WHO grade II) in comparison with controls (6). Microarray analysis coupled with the pharmacologic demethylation of cultured tumor cells is a useful means for investigating genome-wide epigenetic events and has been successfully used for this purpose in glioblastoma; one of the novel candidate genes identified by this approach is AREG (7). CASP8 The CASP8 (caspase 8, apoptosis-related cysteine peptidase 8) gene is located on chromosome 2q33.1. This gene encodes a member of the cysteine-aspartic acid protease (caspase) family. The sequential activation of caspase 8 plays a central role in the execution phase of cell apoptosis (Figure 3.5.1). Caspases exist as inactive proenzymes composed of a prodomain, a large protease subunit, and a small protease subunit. The activation of caspases requires proteolytic processing at conserved internal aspartic residues to generate a heterodimeric enzyme consisting of the large and small subunits. This protein is involved in the programmed cell death induced by Fas and various apoptotic stimuli. The N-terminal FADD-like death effector domain of this protein suggests that it may interact with Fasinteracting protein FADD (61). A significant epigenetic silencing of the proapoptotic CASP8 gene was observed during the progression of primary to recurrent GBM, probably conferring tumor cells a relevant further growth advantage. In brain tumors, CASP8 is hypermethylated in 40% of neuroblastomas and medulloblastomas. Moreover, a high concordance between gene 27 methylation, decreased mRNA levels, and protein expression was observed as well (26). Figure 3.5.1. Apoptotic TRAIL signaling Binding of TRAIL to death receptors (DR4, DR5) leads to the recruitment of the adaptor molecule, FADD. DcR1/2 work as inhibitors of DR4/5. Pro-caspase-8 binds to FADD leading to DISC formation and resulting in its activation. Activated caspase-8 directly activates executioner caspases (caspase-3, -6, and -7) (type I cells) or cleaves Bid (type II cells). Translocation of the truncated Bid (tBid) to the mitochondria promotes the assembly of Bax-Bak oligomers and mitochondria outer membrane permeability changes. Cytochrome c is released into cytosol resulting in apoptosome assembly. Active caspase-9 then propagates a proteolytic cascade of effector caspases activation that leads to morphological hallmarks of apoptosis. Further cleavage of pro-caspase-8 by effector caspases generates a mitochondrial amplification loop that further enhances apoptosis. When FLIP levels are elevated in cells, caspase-8 preferentially recruits FLIP to form a caspase-8-FLIP heterodimer which does not trigger apoptosis (64). A study by Qi et al. showed the heterogeneity of glioblastomas and derived cancer stem cells in the genomic status of CASP8, expression of CASP8, and thus responsiveness to TRAIL-induced apoptosis. Clinic trials may consider genomic analysis of the cancer tissue to identify the genomic 28 loss of CASP8 and use it as a genomic marker to predict the resistance of glioblastomas to TRAIL apoptosis pathway-targeted therapies (62). The loss of CASP8 expression in cancer cells may result in resistance to drug-induced apoptosis and may be important for a therapeutic strategy. Recent studies have demonstrated that the demethylation of CpG islands of the CASP8 gene in resistant cancer cells sensitizes them to chemotherapeutic agents (63). Moreover, the inactivation of CASP8 expression by promoter hypermethylation may result in cancer progression. COX7A1 The COX7A1 (cytochrome c oxidase subunit VIIa polypeptide 1) gene is located on chromosome 19q13.1. The product of this gene is one of the nuclear-coded polypeptide chains of cytochrome c oxidase, the terminal oxidase in mitochondrial electron transport (70). COX catalyses the electron transfer from reduced cytochrome c to oxygen, producing H2O. This reaction is coupled with proton transfer from the matrix to the intermembrane space, thereby contributing to the energy stored in the electrochemical gradient. The catalytic core of COX is composed of mitochondrial encoded proteins whereas nucleus-encoded proteins contribute to the structural and regulatory subunits of the complex. COX7A1 promoter methylation has been correlated with gene expression (71). COX7A1 demonstrated increased expression after 5-azadc treatment in methylated breast tumor cell lines (72). In an independent cohort of breast tumor/normal pairs, COX7A1 demonstrated increased methylation in 70.6% (12/17) of tumors compared with a corresponding normal tissue, and a further 5 showed the equal levels of low-level methylation in a tumor and a corresponding normal tissue (72). This gene has been detected differentially expressed in lung cancer (73). CD81 The CD81 (CD81 molecule, TAPA-1) gene is located on chromosome 11p15.5. This gene is localized in the tumor-suppressor gene region, and thus it is a candidate gene for malignancies (65). It encodes a cellular membrane protein belonging to the tetraspanin family. Most of these members are cell-surface proteins that are characterized by the presence of 4 hydrophobic domains. This encoded protein is a cell surface glycoprotein that is known to form a complex with integrins. The protein complex mediates the events of signal transduction that play a role in the regulation of cell development, activation, growth, and motility. In addition, CD81 29 may be involved in signal transduction. There are data that CD81 tetraspanin may have an inhibitory effect on cell movement and metastases (Figure 3.5.2) (66, 67). The gene was found to be silenced by methylation in multiple myeloma cell lines (68). Recent glioblastoma methylome studies have shown a CD81 methylation rate of 54% (2, 6). Figure 3.5.2. Tetraspanin signaling CD81 (and CD9) is associated with phosphatidylinositol 4-kinase (PI4K), which locally produces phosphoinositides (such as phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2)). This causes the recruitment and activation of SHC (SRC-homology-2domain-containing transforming protein). Subsequent Ras-mediated activation of extracellular signal-regulated kinase (ERK) or p38 or Jun N-terminal kinase (JNK) pathways leads to proliferation or apoptosis, respectively. Caspase-3 activation might also contribute to CD9-dependent regulation of apoptosis. Signaling through CD151 might negatively regulate Ras-ERK/MAPK and Akt/protein kinase B (PKB) signaling in a celladhesion-dependent manner. CD151 (together with associated laminin-binding integrins) also preferentially activates Rac (and Cdc42) over Rho, leading to regulation of the actin cytoskeleton, cell spreading and motility. Conversely, CD82 might suppress tumor cell invasion by downregulating p130Cas and thereby decreasing Rac activation. The Cterminal tails of CD151 (and CD81and CD9) contain PDZ-domain-binding motifs, and the tail of CD151 is essential for its signaling and adhesion-strengthening functions. Possibly, PDZ-domain-containing proteins could connect these tetraspanins to signaling and cytoskeletal regulatory events that affect adhesion strengthening, cell spreading and cell motility (69) . 30 DcR1 Despite 8p being a relatively small chromosome arm, it is one of the most frequently altered genomic regions in human cancer, and is also rich in candidate oncogenes and tumor suppressor genes associated with the development of certain types of cancers (74). Deletions in this region are observed in glioblastoma (75). One of these genes located on chromosome 8p region is DcR1 (tumor necrosis factor receptor superfamily, member 10c, decoy without an intracellular domain, TNFRSF10C). The protein encoded by the DcR1 gene is a member of the TNF-receptor superfamily. DcR1 lacks the intracellular death domain, can bind TRAIL, but is not capable of transmitting the death signal because of its defective death domains (76). This receptor is not capable of inducing apoptosis and is thought to function as an antagonistic receptor that protects cells from TRAIL-induced apoptosis. The expression of this gene was detected in many normal tissues but not in most cancer cell lines, which may explain the specific sensitivity of cancer cells to the apoptosis-inducing activity of TRAIL (77). DcR1 promoter methylation was observed in 40.4% of primary breast tumors (78). Also, high methylation levels at the DcR1 promoter region in peripheral blood was positively associated with perineural tumor spread and shorter patients’ survival (79). DcR1 was frequently hypermethylated in hepatocellular carcinoma tissue samples, and its methylation was found to be closely associated with the inactivation of gene expression (80). As an alternative to a deletion, DcR1 promoter methylation may be associated with clinical data of glioblastoma patients. The methylation of the DcR1 gene promoter was determined in 21% of low-grade gliomas (6) suggesting its role in gliomagenesis. TRAIL acts through 2 pairs of opposing receptors, 2 proapoptotic death receptors, DR4 and DR5, and 2o antiapoptotic decoy receptors, DcR1 and DcR2. All TRAIL receptors have been mapped to the same chromosomal locus, 8p21-22, suggesting evolution from a common ancestral gene (76). DR4 The gene DR4 (TNFRSF10A, tumor necrosis factor receptor superfamily, member 10a) is located on chromosome 8p21. DR4 is a receptor for the cytotoxic ligand TNFSF10/TRAIL. The adapter molecule FADD recruits Casp8 to the activated receptor. The resulting death-inducing signaling complex (DISC) performs Casp8 proteolytic activation, which initiates the subsequent cascade of caspases (aspartate-specific cysteine proteases) mediating apoptosis (81). The methylation of the DR4 gene promoter was found to be extremely common, in both early gastric carcinomas and 31 advanced gastric carcinomas (98.4% and 83.3%, respectively) (76). rhTRAIL and 2 newly designed monoclonal antibodies targeting DR4 (mapatumumab and HGS-ETR1) are new promising cancer drugs because these agents have been shown to preferentially induce apoptosis in tumor cells. However, the main obstacle in TRAIL-based therapies remains the resistance of cancer cells to TRAIL and its analogues. Therefore, identifying the resistance mechanisms and the predictors of response in these tumors remains a key issue in modern cancer diagnostics and therapeutic strategies. Among other possibilities, such predictive factors may be epigenetically regulated in a tumor-specific manner (49). DR4 promoter methylation is frequent in human astrocytic gliomas, and the epigenetic silencing of DR4 mediates resistance to TRAIL/DR4-based glioma therapies (49). The hypermethylation of DR4 (up to 66.7%) was observed in neuroblastomas (82). In non-CNS malignancies, higher methylation rates of DcR1 and DcR2 were reported (up to 100%), whereas DR4 and DR5 were rarely hypermethylated (83). In contrast to the findings reported by van Noesel et al. and Shivapurkar et al., Martinez et al. did not observe the CpG island hypermethylation of the potentially antiapoptotic DcR1 in any GBM as well as the lower rates of hypermethylation of DR4 and DR5 (25 and 12.5%, respectively) (26). The development of targeted therapies restoring functional extrinsic apoptosis, as recently shown in vivo with the synergistic combination of the DNA demethylating agent decitabine and TRAIL, may provide a useful tool to overcome the resistance of GBM to contemporary treatment modalities. GATA4 The GATA4 (GATA binding protein 4) gene is located on chromosome 8p23.1. This gene encodes a member of the GATA family of zinc-finger transcription factors. The members of this family recognize the GATA motif, which is present in the promoters of many genes (84). GATA4 regulates the gene expression of various genes, which are involved in cardiac development, cardiac hypertrophy, and heart failure. In addition to the heart, GATA4 plays important roles in the reproductive system, gastrointestinal system, respiratory system, and cancer. Therefore, the positive and negative regulations of GATA4 are important components of biologic functions (85). Recently, GATA4 expression in normal embryonic and adult mouse and human astrocytes has been reported, in which it functions as an inhibitor of proliferation and inducer of apoptosis (86). GATA4 knock-out in mice lead to embryonic lethality due to cardiac defects (87, 88), and GATA4 mutations cause Holt-Oram syndrome and congenital 32 heart defects. GATA4 is frequently silenced in lung, colon, prostate, ovarian, and breast cancer (89-93), but its exact role in cancer biology and the mechanisms by which it operates are poorly understood (94). The expression of GATA4 was extinguished in the majority of cell lines from colorectal and gastric cancers as well as in primary tumors. Silencing was associated with the hypermethylation of the GATA4 promoter sequences. Moreover, the candidate target genes of the GATA4 factor were commensurately decreased in their expression and also displayed the methylated promoters (95). GATA4 loss occurred through promoter hypermethylation or novel somatic mutations (94). The re-expression of GATA4 also conferred the sensitivity of GBM cells to temozolomide, a DNA alkylating agent currently used in the GBM therapy (94). GATA4 can be activated through stimulating the activity of the already existing GATA4 protein molecule or by increasing the GATA4 protein expression. GATA4 protein expression can be altered through transcriptional or translational mechanisms (85). Given the role of GATA4 in regulating astrocyte proliferation and the observed loss of GATA4 in several human cancers, GATA4 was demonstrated to be a novel tumor suppressor in GBM. GATA6 The GATA6 (GATA binding protein 6) gene is located on chromosome 18q11.2. GATA6 is one of 6 members of the mammalian GATA family of transcription factors, all containing 2 highly conserved zinc-finger DNA binding domains that interact with a canonical DNA motif (G/A)GATA(A/T) (96). It is thought that GATA6 may be important for regulating terminal differentiation and/or proliferation. It is also linked to embryonic cardiac development. The loss of GATA6 resulted in the enhanced proliferation and transformation of astrocytes (52). Knockin GATA6 expression in human malignant astrocytoma cells reduced their tumorigenic growth with decreased VEGF expression. The disruption of GATA6 led to transformation and demonstrated that GATA6 is a new and relevant human GBM tumor suppressor gene (52). The great majority of human glioblastomas (which can arise from low-grade astrocytomas), but not low-grade astrocytomas, displayed the loss of GATA6 expression, mutations in GATA6, and loss of heterozygosity. The re-expression of GATA6 in human malignant astrocytoma cells inhibited their growth (52). This establishes GATA6 as a bona fide tumor suppressor in this disease, marking the progression from low-grade astrocytoma to malignant glioblastoma (95). 33 hMLH1 The hMLH1 (MutL homolog 1, colon cancer, nonpolyposis type 2 E. coli) gene is a human gene located on chromosome 3p21.3. hMLH1 plays a role in DNA mismatch repair. hMLH1 heterodimerizes with PMS2 to form MutL alpha, a component of the postreplicative DNA mismatch repair system (MMR) (Figure 3.5.3) (97). Figure 3.5.3 Mismatch repair A mispaired base is recognized by the hMSH2/GTBP complex while an insertion/deletion loop is recognized by the hMSH2/hMSH3 complex. MutL related proteins (hMLH1/hPMS2 and hMLH1/hPMS1 complexes) then interact with the MutS related proteins that are already bound to the mispaired bases. The hMSH2/GTBP complex may also support the repair of insertion/deletion loops (97). DNA repair is initiated by MutS alpha (MSH2-MSH6) or MutS beta (MSH2-MSH6) binding to a dsDNA mismatch, and then MutL alpha is recruited to the heteroduplex. The assembly of the MutL-MutS34 heteroduplex ternary complex in the presence of RFC and PCNA is sufficient to activate the endonuclease activity of PMS2. It introduces single-strand breaks near the mismatch and thus generates new entry points for the exonuclease EXO1 to degrade the strand containing the mismatch. DNA methylation would prevent cleavage and therefore assure that only the newly mutated DNA strand is going to be corrected. MutL alpha (MLH1PMS2) interacts physically with the clamp loader subunits of DNA polymerase III, suggesting that it may play a role to recruit DNA polymerase III to the site of the MMR. Also implicated in DNA damage signaling, a process, which induces cell cycle arrest and can lead to apoptosis in case of major DNA damages. hMLH1 heterodimerizes with MLH3 to form MutL gamma, which plays a role in meiosis (98). hMLH1 inactivation by promoter hypermethylation has been reported to be associated with some human cancers (99-101). Herman et al. have suggested that DNA methylation associated with the transcriptional silencing of hMLH1 is the underlying cause of mismatch repair defects in most sporadic colorectal cancers (102). Xinarianos et al. have shown that 58.6% and 57.8% of lung cancer tumor specimens had the reduced expression levels of the hMLH1 and hMSH2 proteins, respectively (103). KRT81 The KRT81 (keratin 81, also KRTHB1 or Hb-1) gene is located on chromosome 12q13. The protein encoded by this gene is a member of the keratin gene family. As type II hair keratin, it is a basic protein that heterodimerizes with type I keratins to form hair and nails (104). Keratins are proteins expressed in all types of epithelial cells (105), with different expression patterns among different carcinomas (106), and they are extensively used as diagnostic markers. To date, KRT81 has not been validated as a prognostic marker, and the present study is the first to link a KRT81 variant to tumor recurrence. Campayo et al. have found that KRT81 may be a new immunohistochemical marker of squamous cell carcinoma (107). KRT81 showed a clear positive staining in squamous cell carcinomas (95% positive), whereas 81% of adenocarcinomas were negative (107). Their findings indicate that single nucleotide polymorphisms in KRT81 and XPO5 could be proved to be useful biomarkers for individualizing the therapy in patients with non–small cell lung cancer and that KRT81 may be a novel immunohistochemical marker of squamous cell carcinoma, providing a new diagnostic tool to be used in therapeutic decision-making (107). 35 NPTX2 The NPTX2 (neuronal pentraxin II) gene is located on chromosome 7q22.1. NPTX2 gene encodes a member of the family of neuronal pentraxins, synaptic proteins that are related to C-reactive protein. This protein is involved in excitatory synapse formation. It also plays a role in the clustering of AMPA-type glutamate receptors at established synapses, resulting in nonapoptotic cell death of dopaminergic nerve cells. Upregulation of this gene in the tissues of patients with Parkinson disease suggests that the protein may be involved in the pathology of Parkinson disease (108). Its methylation status was recently shown to be correlated with age (109, 110). The aberrant methylation of NPTX2 was also detected in 98% of primary pancreatic cancers, and it was not methylated in any of the normal ductal epithelia studied (111). In vitro data showed that NPTX2 suppressed tumor cell proliferation, migration, and invasion. In addition, experiments showed that NPTX2 affected cell cycle and apoptosis indicating that NPTX2 might be a promising candidate tumor suppressor gene (112). NPTX2 methylation has been demonstrated in various types of cancers. The methylation frequencies of NPTX2 were statistically higher in the malignant cholangiocarcinoma and pancreatic carcinoma group (54.55%) than in the benign group (27.78%) (113). NPTX2 appeared to be 8% methylated in primary Mantle cell lymphoma. Furthermore, the number of Ki-67 positive cells correlated significantly with the methylation levels of NPTX2 in primary Mantle cell lymphoma (114). Although the methylation status of NPTX2 showed a tendency to be associated with shorter overall survival, it did not reach statistical significance (114). Mueller et al. in 2006 have suggested that NTPX2 is probably silencing by methylation in glioblastomas (7). SPINT1 The SPINT1 (serine peptidase inhibitor, Kunitz type 1, HAI-1, hepatocyte growth factor activator inhibitor type 1) gene is located on chromosome 15q15.1. SPINT1 strongly inhibits HGFAC, trypsin, KLK4, KLK5, matriptase/ST14, prostasin/PRSS8, and hepsin/TMPRSS1. SPINT1 seems to have tumor suppressor activity: transgenic overexpression of matriptase/ST14 resulted in skin carcinogenesis. However, the development of skin cancer (squamous cell carcinoma) was suppressed when SPINT1 was coexpressed (115). 36 TES The TES (testis derived transcript, 3 LIM domains) gene, also known as testing, is located on chromosome 7q31.2. This gene is similar to mouse testin, a testosterone-responsive gene encoding a Sertoli cell secretory protein containing 3 LIM domains. LIM domains are double zincfinger motifs that mediate protein-protein interactions between transcription factors, cytoskeletal proteins, and signaling proteins. This protein is a negative regulator of cell growth and may act as a tumor suppressor. This scaffold protein may also play a role in cell adhesion, cell spreading, and the reorganization of the actin cytoskeleton. Multiple protein isoforms are encoded by transcript variants of this gene (116). The loss of TES expression also occurs frequently in various cancers (117, 118). Missense mutations are scarce, and homozygous deletions have not been observed, consistent with CpG promoter hypermethylation being a mechanism of TES gene inactivation (118). Forced TES expression in HeLa or OVCAR5 cells profoundly reduces cancer cell growth (118). TES knockout mice showed an increased susceptibility to carcinogen-induced (nitrosomethylbenzylamine) gastric cancer (119). In addition, the restoration of TES by adenoviral transduction of non-TES expressing breast cancer and uterine sarcoma cell lines inhibited their growth by the induction of apoptosis (120). Frequent promoter hypermethylation in a subset of glioblastomas is therefore consistent with the possibility of similar tumor-related inactivation in highgrade gliomas (7). TES methylation has been shown in primary tumors, including glioblastomas (7) and ovarian cancer (118). Methylation at a single site in the TES promoter has been reported for several cell lines including lymphoid leukemia, breast cancer and pancreatic cancer cells (117). TES methylation is closely associated with the loss of TES expression in cell lines (117) and glioblastoma cells (7). A reciprocal relationship between TES promoter methylation and expression has been demonstrated (121). TFPI-2 The TFPI-2 (tissue factor pathway inhibitor 2) gene is located on chromosome 7q22. It may play a role in the regulation of plasmin-mediated matrix remodeling. It inhibits trypsin, plasmin, factor VIIa/tissue factor, and weakly factor Xa (122). TFPI2 has also been described as a MMP inhibitor. MMP by effecting the proteolytic degradation or activation of cell surface and extracellular matrix (ECM) proteins can modulate both cell-cell and cell-ECM interactions, which influence cell differentiation, migration, 37 proliferation, and survival (123). The matrix metalloproteinase inhibitor TIMP-2 has a high specificity for gelatinase A/MMP-2 (124). An imbalance between MMP and TIMP-2 in favor of enzymatic activity is linked to the degradation of the extracellular matrix associated with several physiologic and pathologic events, including angiogenesis, invasion, and metastasis. Since TIMPs are secreted molecules, they have the potential to be used for the gene therapy of certain tumors (124). TFPI-2 was found to be abundantly expressed in various normal human tissues, including seminal vesicles, colon, stomach, brain, pancreas, esophagus, and liver (125-128). Several studies suggested that the TFPI-2 gene is inactivated or absent during tumor progression. The expression of TFPI-2 diminishes with an increasing degree of malignancy, which may suggest a role for TFPI-2 in the maintenance of tumor stability and inhibition of the growth of neoplasms (126). Besides gene locus deletion and aberrant splicing, the mechanism responsible for TFPI-2 down-regulation in tumor cells has been majorly attributed to promoter hypermethylation (129). Transcriptional silencing by the promoter hypermethylation of TFPI-2 was observed in some human cancers (129). Gessler et al. have suggested that the antiinvasive properties of TFPI-2 are associated with inhibition of MMP-1 and MMP-2, while the inhibition of MMP-9 seems to play a minor role in this context (130). Their findings underscore the important role of TFPI-2 as a tumor suppressor gene and indicate that TFPI-2 may be a useful diagnostic marker for the aggressive phenotype of glial tumors (130). 38 4. MATERIALS AND METHODS 4.1. Patient tumor samples and characteristics Part of the study was done with 22 glioblastoma samples obtained from patients who underwent surgery in the Department of Neurosurgery, Charité, Campus Virchow Klinikum (Berlin, Germany), after ethics committee approval and patient consent. Glioblastoma samples were snap frozen. All glioblastomas were histologically diagnosed according to the WHO criteria. These samples were used for the analysis of COX7A1, SPINT1, AREG, NPTX2, and KRT81 gene expression and methylation. However, there were no data available on the clinical characteristics of these patients. Another part of the study included 100 WHO grade IV glioblastomas. All glioblastomas were surgically resected and histologically diagnosed according to the 2007 WHO criteria (131) in the Clinic of Neurosurgery, Hospital of Lithuanian University of Health Sciences, Kaunas, Lithuania, between 2003 and 2009. The date of database closure was November 2010. The tissue samples were coded and separated from patients’ names and personal information prior to their collection. Tumor samples were collected, following written informed consent, in accordance with the Lithuanian regulations and the Helsinki Declaration. Written informed consent was obtained for every patient under the approval of the Ethics Committee, Lithuanian University of Health Sciences. The following clinical data were recorded for each patient: age at the time of the operation, gender, time of the last follow-up, and patient status. Overall survival times were recorded for all cases and were calculated from the time of operation to death or last contact with live patients. 4.2. RNA extraction Total RNA and DNA from glioblastoma tissue cells was purified using a TRIZOL reagent (Invitrogen, Carlsband, CA, USA) following the manufacturer’s recommendation. Genomic RNA and DNA were isolated from glioblastoma tissue samples by crushing frozen samples with a mortar and pestle. 39 Reagents: TRIzol Reagent (Invitrogen) Ethanol (Fisher) Isopropanol (Fisher) Chloroform (Fisher) 75% ethanol (in DEPC-treated water) RNase-free water Equipment: 10/100/1000 µL single-channel pipettes (Eppendorf) Homogenizer (Kinematica Hand Held Polytron) Spectrophotometer (Nanodrop) RNAse-free 1.5 mL tubes (Ambion) Centrifuge 5415 R (Eppendorf AG) Protocol: 1. Add 0.75 mL of TRIzol LS Reagent per 50-100 mg of tissue sample and homogenize. Incubate the homogenized sample for 5 minutes at room temperature to permit complete dissociation of the nucleoprotein complex. 2. Add 0.2 mL of chloroform per 0.75 mL of TRIzol LS Reagent used for homogenization. Cap the tube securely. Shake tube vigorously by hand for 15 seconds. Incubate for 2-15 minutes at room temperature. 3. Centrifuge the sample at 12 000g for 15 minutes at 4°C. Note: The mixture separates into a lower red phenol-chloroform phase, an interphase, and a colorless upper aqueous phase. RNA remains exclusively in the aqueous phase. The upper aqueous phase is ~70% of the initial volume of TRIzol LS Reagent used for homogenization. 4. Remove the aqueous phase of the sample by angling the tube at 45° and pipetting the solution out. Avoid drawing any of the interphase or organic layer into the pipette when removing the aqueous phase. 5. Place the aqueous phase into a new tube and proceed to the RNA Isolation Procedure. (Save the interphase and organic phenolchloroform phase if isolation of DNA or protein is desired. See DNA Isolation Procedure and Protein Isolation Procedure for details. The organic phase can be stored at –70°C). 6. Add 0.5 mL of 100% isopropanol to the aqueous phase, for every 0.75 mL TRIzol LS Reagent used for homogenization. Incubate at room temperature for 10 minutes. 40 7. Centrifuge at 12 000g for 10 minutes at 4°C. (The RNA is often invisible prior to centrifugation, and forms a gel-like pellet on the side and bottom of the tube). 8. Remove the supernatant from the tube, leaving only the RNA pellet. 9. Wash the RNA pellet, with 1 mL of 75% ethanol per 0.75 mL of TRIzol LS Reagent used for the initial homogenization. Vortex the sample to mix. 10. Centrifuge the sample at 7500g for 5 minutes at 4°C and discard the supernatant. Air dry the RNA pellet for 5-10 minutes. 11. Resuspend the RNA pellet in RNase-free water by passing the solution up and down several times through a pipette tip. 12. Store at –70°C. Analyze the RNA on a spectrophotometer (Nanodrop). A260/280 ratios should be between 1.8 and 2.1. A260/230 ratios should be at least 1.9, more than 2.0 is preferable. A260/280 ratios outside this range indicate DNA or protein contamination. Low A260/280 ratios indicate carbohydrate, phenol, salt contamination. 4.3. DNA extraction DNA extraction from glioblastoma tissue samples using TRIZOL Reagents: Ethanol (Fisher) 75% ethanol (in DEPC-treated water) 0 1 M sodium citrate in 10% ethanol DNase-free water Equipment: 10/100/1000 µL single-channel pipettes (Eppendorf) Centrifuge 5415 R (Eppendorf AG) Spectrophotometer (Nanodrop) Protocol: DNA is isolated from the interphase and phenol-chloroform layer saved from the RNA phase separation step: 1. Remove any remaining aqueous phase overlying the interphase. This is critical for the quality of the isolated DNA. 41 2. Add 0.3 mL of 100% ethanol per 0.75 mL of TRIzol LS Reagent used for the initial homogenization. Cap the tube and invert the sample several times to mix. Incubate samples for 2-3 minutes at room temperature. 3. Centrifuge the tube at 2000g for 5 minutes at 4°C to pellet the DNA. Remove the phenol-ethanol supernatant and save it in a new tube if protein isolation is desired. The supernatant can be stored at –70°C for several months. 4. Wash the DNA pellet with 1 mL of sodium citrate/ethanol solution (0.1 M sodium citrate in 10% ethanol, pH 8.5) per 0.75 mL of TRIzol® LS Reagent used for the initial homogenization. Incubate for 30 minutes at room temperature. Mix occasionally by gentle inversion. 5. Centrifuge at 2000g for 5 minutes at 4°C. Remove and discard supernatant. 6. Repeat wash (steps 4-5), once. 7. Add 1.5-2 mL of 75% ethanol per 0.75 mL TRIzol LS Reagent used for the initial homogenization. Incubate for 10-20 minutes at room temperature. Mix the tube occasionally by gentle inversion. 8. Centrifuge at 2000g for 5 minutes at 4°C. Remove and discard supernatant. 9. Air dry the DNA pellet for 5-10 minutes. Do not allow the pellet to dry out. 10. Resuspend the DNA in 50 µL of water. 11. The DNA can be stored at –20°C. Analyze the DNA on a spectrophotometer (Nanodrop). A260/280 ratios should be between 1.6 and 1.8 DNA extraction from glioblastoma tissue samples using ZR Genomic DNA™ Tissue MiniPrep Tumor DNA was extracted from 25-40 mg of frozen tissue using ZR Genomic DNA™ Tissue MiniPrep (Zymo Research, USA) according to manufacturer’s protocol. DNA was isolated from glioblastoma tissue samples by crushing frozen samples with a mortar and a pestle. Reagents: ZR Genomic DNA™ Tissue MiniPrep Kit (Zymo Research, USA) Beta-mercaptoethanol 42 Equipment: 10/100/1000 µL single-channel pipettes (Eppendorf) 1.5 mL tubes (Eppendorf) Centrifuge 5415 R (Eppendorf AG) Thermomixer comfort (Eppendorf AG) MS2 Minishaker (IKA, Wilmington, USA) BioPhotometer (Eppendorf AG) Reagent preparation: • Add 260 μL Proteinase K Storage Buffer to each Proteinase K tube prior to use. The final concentration of Proteinase K after the addition of Proteinase K Storage Buffer is ~20 mg/mL. •Add beta-mercaptoethanol to the Genomic Lysis Buffer to a final dilution of 0.5% (v/v) i.e. Protocol: 1. To a tissue sample (≤25 mg) in a microcentrifuge tube add a solution of: 95 μL of H2O 95 μL of 2× digestion buffer 10 μL of proteinase K 2. Mix and then incubate the tube at 55ºC overnight. 3. Add 700-μL genomic lysis buffer mix thoroughly by vortexing. Centrifuge at 10 000 g for one minute to remove insoluble debris. 4. Transfer the supernatant to a Zymo-Spin™ IIC Column in a collection tube and centrifuge at 10 000g for one minute. 5. Add 200 μL of DNA pre-wash buffer and centrifuge at 10 000g for one minute. 6. Add 400 μL of g-DNA wash buffer to the spin column and centrifuge at 10 000g for one minute. 7. Add 50 μL of water to the spin column. Incubate for 2-5 minutes at room temperature and then centrifuge at top speed for 30 seconds to elute the DNA. The eluted DNA can be stored –20ºC for future use. 43 4.5. cDNA synthesis First-Strand cDNA Synthesis Using SuperScript™ II RT Invitrogen Equipment: 10/100 µL single-channel pipettes (Eppendorf) Centrifuge 5415 R (Eppendorf AG) Spectrophotometer (Nanodrop) Thermomixer comfort (Eppendorf AG) MS2 Minishaker (IKA, Wilmington, USA) RNAse-free 0.5 mL tubes (Ambion) Ice bath Protocol: • Add the following components to a nuclease-free microcentrifuge tube: RNA 5 µg 10 mM dNTP mix 1 µL Oligo(dT)12-18 (0.5 µg/µL) 1 µL DEPC- water to 10 µL • Heat mixture to 65°C for 5 minutes and quick chill on ice. Collect the contents of the tube by brief centrifugation and add: 2 µL 4 µL 2 µL 2 µL 10x RT buffer 25 mM MgCl2 0.1 M DTT RNase inhibitor • • • • • Mix contents of the tube gently and incubate at 42°C for 2 minutes. Add 1 μL of SuperScript™ II RT and mix by pipetting gently up and down. Incubate at 42°C for 50 minutes. Inactivate the reaction by heating at 70°C for 15 minutes. To remove RNA complementary to the cDNA, add 1 μL (2 units) of RNase H, and incubate at 37°C for 20 minutes. cDNA amplification cDNA amplification reaction was performed in a total volume of 10 µL, using AmpliTaq Gold PCR Master Mix (Applied Biosystems, USA) and 10 pmol of each primer (Metabion International AG, Germany). 44 Equipment: MS2 Minishaker (IKA, Wilmington, USA) Centrifuge 5415 R (Eppendorf AG, Germany) Mastercycler epgradient S (Eppendorf AG, Germany) 10/100 µL single-channel pipettes (Eppendorf) Protocol for one sample: 2x AmpliTaq Gold PCR Master Mix Primer I PrimerII Water cDNA Total volume 5 µL 0.5 µL 0.5 µL 3 µL 1 µL 10 µL cDNA amplification reaction conditions and cDNA PCR primers are shown in Tables 4.5.1 and 4.5.2. Table 4.5.1. cDNA amplification reaction conditions Step Initial DNA denaturation DNA denaturation Annealing Extension Final Extension END Temperature, ºC Duration 95 95 See table below 72 72 4 5 min 30 s 40 s 30 s 10 min – The number of cycles – 33 – – Table 4.5.2. cDNA PCR primers Primer name NPTXII_cDNAf NPTXII_cDNAr KRTHB1_cDNAf KRTHB1_cDNAr COX7A1_cDNAf COX7A1_cDNAr AREG_cDNAf AREG_cDNAr SPINT1_cDNAf SPINT1_cDNAr GAPDH_cDNAf GAPDH_cDNAr Sequence CAACGACAAGGTTGCGCAGC ACGGTGTCCTGCTCTTGTCC ATGCATCACCACCGTGTCGG GCGCACCTTGTCGATGAAGG AGGACAAGGCAGAATGCAGG GGATGTCATTGTCCTCCTGG TGTCCCAGAGACCGAGTTGC GCATAATGGCCTGAGCCGAG CATCAACTGCCTCTACGAGC ATCCCAGACCCTCCAAAGCC GGTTTTTCTAGACGGCAGGTCA TGGCAAATTCCATGGCACCGTCA 45 Tm, ºC 63 63 63 63 63 63 63 63 58 58 58 58 4.6. DNA bisulphite modification DNA Bisulphite modification using the CpGenome DNA Modification Kit Sodium bisulphite modification is based on the selective demetilation of unmethylated cytosines to uracils where methylated cytosines remain unchanged. This chemical reaction converts a difference in methylation into a difference in sequence. Genomic DNA (1 µg) was modified with sodium bisulphite using the CpGenome DNA Modification Kit (Chemicon International) according to the manufacturer’s instructions. Reagents: CpGenorneTDmN A Modification Kit β-Mercaptoethanol Ethanol EDTA Tris Base NaOH pellets (Chemicon International) (Sigma) (Fishers) (Metford) (Fishers) Equipment: 10/100 µL single-channel pipettes (Eppendorf) Centrifuge 5415 R (Eppendorf AG) Spectrophotometer (Nanodrop) Thermomixer comfort (Eppendorf AG) MS2 Minishaker (IKA, Wilmington, USA) Microcentrifuge tubes, 1.5 mL (Eppendorf) pH electrode Protocol: Reagent Preparation 1. 3 M NaOH stock (freshly prepared prior to each use): dissolve 1 g of dry NaOH pellets in 8.3 mL of water. 2. 20 mM NaOH/90% ethanol (freshly prepared prior to each use). To prepare 1 mL of this solution, combine 900 μL of 100% ethanol, 93.4 μL of H2O, and 6.6 μL of 3 M NaOH. 3. Dissolve Reagent I (freshly prepared prior to each use). Warm bottle to room temperature before opening. For each sample to be modified, weigh 0.227 g of DNA Modification Reagent I and add to 0.571 mL of water. Mix thoroughly by vortexing. Adjust the pH to 46 5.0 with approximately 20 μL of 3M NaOH. Protect the Reagent I solution from light. 4. Dissolve Reagent II. Warm bottle to room temperature before opening. Add 1 μL of -mercaptoethanol to 20 mL of deionized water. Add 750 μL of this solution to 1.35 g of DNA Modification II for each sample to be modified. Mix well to ensure complete dissolution. DNA Modification Procedure: 1. In screw cap 1.5-2.0 mL microcentrifuge tubes: to 1 μg of DNA, add 2 μL of DNA Modification Reagent IV to the sample DNA and bring the total volume to100 μL with water. Then add 7.0 μL of 3M NaOH and mix. 2. Incubate DNA for 10 minutes at 50°C (heat block or water bath). 3. Add 550 μL of freshly prepared DNA Modification Reagent I and vortex. 4. Incubate at 50°C for 4-16 hours in a heat block or a water bath protected from light. 5. Resuspend DNA Modification Reagent III by vortexing vigorously. Draw suspension into and out of a 1 mL plastic pipette tip 10× to disperse any remaining clumps. 6. Add 5 μL of well-suspended DNA Modification Reagent III to the DNA solutions in the tubes. 7. Add 750 μL of DNA Modification Reagent II and mix briefly. 8. Incubate at room temperature for 5-10 minutes. 9. Spin for 10 seconds at 5000g to pellet the DNA Reagent III (a small white pellet should be present). Discard supernatant. 10. Add 1.0 mL of 70% EtOH, vortex and spin for 10 seconds at 5000g, and discard supernatant. Perform this step 3 times. 11. After the supernatant from the third wash has been removed, centrifuge the tube at high speed for 2 minutes, and remove the remaining supernatant with a plastic pipette tip. 12. Add 50 μL of the 20 mM NaOH/90% EtOH solution to the appropriate samples. 13. Vortex briefly to resuspend the pellet, and incubate at room temperature for 5 minutes. 14. Spin for 10 seconds at 5000g to move all contents to the tip of a tube. Add 1.0 mL of 90% ethanol and vortex to wash the pellet. Spin again and remove the supernatant. Repeat this step one more time. 15. After the supernatant from the second wash has been removed, centrifuge the sample at high speed for 3 minutes. 47 16. Remove all of the remaining supernatant with a plastic pipette tip. Allow the tube to dry for 10-20 minutes at room temperature (alcohol odor should diminish). 17. Add 25 μL of TE Buffer. 18. Incubate the sample for 15 minutes at 50°C-60°C to elute DNA. 19. Centrifuge at high speed for 2-3 minutes and transfer the sample (supernatant) to a new tube using a plastic pipette tip. 20. Proceed to MSP or sequencing, or store at –15°C to –25°C for up to 2 months, at –80°C for up to 6 months. Bisulfite conversation of DNA using EZ DNA Methylation Kit The methylation status of genes promoters was determined by bisulfite treatment of DNA. An amount of 400 ng of DNA was used for bisulfite modification. DNA modification was performed using EZ DNA Methylation Kit (Zymo Research, USA), and all procedures were done according to the manufacturer’s protocol. Equipment: 10/100 µL single-channel pipettes (Eppendorf) Centrifuge 5415 R (Eppendorf AG) Spectrophotometer (Nanodrop) Thermomixer comfort (Eppendorf AG) MS2 Minishaker (IKA, Wilmington, USA) Microcentrifuge tubes, 1.5 mL (Eppendorf) Protocol: Reagent Preparation 1. Preparation of CT Conversion Reagent. Add 750 μL of water and 210 μL of M-Dilution Buffer to a tube of CT Conversion Reagent. Mix at room temperature with frequent vortexing or shaking for 10 minutes. 2. Preparation of M-Wash Buffer. Add 24 mL of 100% ethanol to 6 mL of M-Wash Buffer concentrate. DNA Modification Procedure: 1. Add 5 μL of M-Dilution Buffer to the DNA sample and adjust the total volume to 50 μL with water. Mix the sample by flicking or pipetting up and down. 2. Incubate the sample at 37°C for 15 minutes. 48 3. After the above incubation, add 100 μL of the prepared CT Conversion Reagent to each sample and mix. 4. Incubate the sample in the dark at 50°C for 12-16 hours. 5. Incubate the sample at 0°C-4°C for 10 minutes. 6. Add 400 μL of M-Binding Buffer to a Zymo-Spin™ IC Column and place the column into a provided collection tube. 7. Load the sample (from Step 5) into the Zymo-Spin™ IC Column containing the M-Binding Buffer. Close the cap and mix by inverting the column several times. 8. Centrifuge at full speed (>10 000g) for 30 seconds. Discard the flowthrough. 9. Add 100 μL of M-Wash Buffer to the column. Centrifuge at full speed for 30 seconds. 10. Add 200 μL of M-Desulphonation Buffer to the column and let stand at room temperature (20°C-30°C) for 15-20 minutes. After the incubation, centrifuge at full speed for 30 seconds. 11. Add 200 μL of M-Wash Buffer to the column. Centrifuge at full speed for 30 seconds. Add another 200 μL of M-Wash Buffer and centrifuge for an additional 30 seconds. 12. Place the column into a 1.5 ml microcentrifuge tube. Add 40 μL of MElution Buffer directly to the column matrix. Centrifuge for 30 seconds at full speed to elute the DNA. DNA is ready for immediate analysis or can be stored at or below –20°C for later use. For long-term storage, store at or below –70°C. 4.7. Sodium bisulphite sequencing Sodium bisulphite sequencing is a genomic sequencing technique (132) allowing the generation of methylation maps with single nucleotide resolution based on bisulphite modification of DNA and sequencing of PCR products. The method involves the subcloning of a PCR product into an appropriate vector and sequencing of the inserts of several individual clones. The resulting final sequence pattern shows that all original cytosines appear as thymines, whereas methylated cytosine bases are displayed as cytosines, resulting in the quantification of CGI methylation at every CpG position. 49 Preparing for sequencing using TOPO TA Cloning Kit Protocol for one sample: Component 2x AmpliTaq Gold PCR Master Mix Primer I Primer II Water DNA Total volume Volume 10 μL 1 μL 1 μL 6 μL 2 μL 20 μL cDNA amplification reaction conditions and PCR primers are shown in Tables 4.7.1 and 4.7.2. Table 4.7.1. DNA amplification reaction conditions Step Initial DNA denaturation DNA denaturation Annealing Extension Final Extension END Temperature, ºC 95 95 See table below 72 72 4 Duration 5 min 30 s 60 s 2 min 10 min – No. of cycles – 36 – – Table 4.7.2. PCR primers Primer name NTPX2_pr_bm_Fr_1f NTPX2_pr_bm _Fr_1r NTPX2_pr_bm_Fr_2f NTPX2_pr_bm_Fr_2r NTPX2_pr_bm_Fr_3f NTPX2_pr_bm_Fr_3r KRT81_pr_bm_Fr_1f KRT81_pr_bm_Fr_1r KRT81_pr_bm_Fr_2f KRT81_pr_bm_Fr_2r Cox_pr_bm_Fr_1f Cox_pr_bm_Fr_1r Cox_pr_bm_Fr_2f Cox_pr_bm_Fr_2r SPINT1_pr_bm_Fr_1f SPINT1_pr_bm_Fr_1r AREG_pr_bm__Fr_1f AREG_pr_bm_Fr_1r Sequence TTTAGGTTTTGTTGAATAAGGTTAT TAAAAAAAAAATAAAAACCCCT TTTTTTTTGAGGTAGAGTT CCCTTTCTTAAACTAAAAAAC GTTTTTTAGTTTAAGAAAGGG AACTATCCTAAACCCCAAC GGTGTTTTGAGGAGTATATTGGAGT TAATAATACATAAAAAACTAAACCC GGGTTTAGTTTTTTATGTATTATTAT TATAATCCAAAACACCCACCTTATC TTTGTAAAAATGTATTTTTTGGTAT ACCTACATTCTACCTTATCCTCTTCC GTGAGGTTTTTATGGGTTGGGT AAAAAAATTTCCTAAATTAAAAAAAA TTTTAGGTTTGGGGTTGGGAAAGTA CCTAATTTCTAATAAAACTAAATCAA AGGTTTAAGTTTTATTTTTTTT AAACTCTCATTAATCCTTC 50 Tm, ºC 56 56 47 47 51 51 55 55 55 55 51 51 53 53 60 60 49 49 Remove 10 μL from the reaction and analyze by agarose gel electrophoresis. Prepare the obtained PCR products to clone with TOPO TA Cloning Kit (Invitrogen) according to the manufacturer’s recommendations. TOPO TA Cloning Reaction • Protocol for TOPO Cloning reactions: Reagent Volume Fresh PCR Product 2 μL Salt solution 1 μL Water 2 μL TOPO vector 1 μL • • Mix gently and incubate for 5 minutes at room temperature. Place the tubes on ice. One shot chemical transformation Supplied Materials: • S.O.C. medium (included with the kit) • LB plates containing 50 μg/mL of ampicillin • 40 mg/mL X-gal in dimethylformamide (DMF) 1. Add 2 μL of the TOPO® Cloning reaction into a vial of One Shot® Chemically Competent E. coli and mix gently. 2. Incubate on ice for 30 minutes. 3. Heat-shock the cells for 30 seconds at 42°C without shaking. 4. Immediately transfer the tubes on ice. 5. Add 250 μL of room temperature S.O.C. medium. 6. Cap the tube tightly and shake the tube horizontally (200 rpm) at 37°C for 1 hour. 7. Spread 10-50 μL from each transformation on a prewarmed selective plate. 8. Incubate plates at 37°C overnight. 9. Pick ~10 white or light blue colonies for analysis. Do not pick dark blue colonies. LB-medium agar-plates (1 L): 1. Dissolve 10 g of tryptone, 5 g of yeast extracts, 15 g of agar, and 10 g og NaCl in 950 mL of deionized water. 51 2. Adjust the pH of the medium to 7.0 using 1 N NaOH and bring volume up to 1 liter. 3. Autoclave on liquid cycle for 20 minutes at 15 psi. Allow solution to cool to 55°C and add antibiotic (50 µg/mL of Amp or Kan) and pour into Petri dishes. 4. Let harden, then invert, and store at 4°C in the dark. Primers of bacterial plasmids for PCR reaction and cDNA amplification reaction conditions are shown in Tables 4.7.3 and 4.7.4. Table 4.7.3. Primers of bacterial plasmids for PCR reaction Primer M13 Forward M13 Reverse Sequence 5'-GTAAAACGACGGCCAG-3' 5'-CAGGAAACAGCTATGAC-3' Component 2x polymerase mix Primer I Primer II Water Take a little amount of bacterial colonies Total pMoles Supplied 407 pM 385 pM Volume 10 μL 1 μL 1 μL 8 μL 20 μL Table 4.7.4. DNA amplification reaction conditions Step Initial denaturation DNR denaturation Annealing Extension Final extension END Temperature, ºC 95 95 55 72 72 4 Duration 5 min 30 s 1 min 1 min 10 min – Cycles – 25 – – Remove 10 μL from the reaction and analyze by agarose gel electrophoresis. PCR Product Cleanup ExoSAP-IT reagent (Affymetrix, USA) was used for PCR Product Cleanup. It treats PCR products ranging in size from less than 100 bp to more than 20 kpb with absolutely no sample loss by removing unused primers and nucleotides. 52 Protocol: 1. Remove the ExoSAP-IT reagent from a –20°C freezer and keep on ice throughout this procedure. 2. Mix 10 μL of a post-PCR reaction product with 4 μL of ExoSAP-IT reagent for a combined 14-μL reaction volume. 3. Incubate at 37°C for 15 minutes to degrade remaining primers and nucleotides. 4. Incubate at 80°C for 15 minutes to inactivate ExoSAP-IT reagent. The PCR product is now ready for use in DNA sequencing, Treated PCR products may be stored at –20°C until required. Preparation for sequencing using SEQ USA): Component Clean PCR product ABI 5x sequencing buffer Primer M13 (5 pmoL/μL) BDT premix Mili-Q water Total Sequencing Reaction Kit (Montage, Volume 0.5 μL 1.5 μL 0.5 μL 1 μL 6.5 μL 10 μL DNA amplification reaction conditions are shown in Table 4.7.5. Table 4.7.5. DNA amplification reaction conditions Step Initial DNA denaturation DNA denaturation Annealing Extension Final extension Temperature, ºC 93 96 55 60 10 Duration 3s 10 s 15 s 4 min – Cycles – 24 – 4.8. Methylation-specific PCR MSP is a qualitative PCR-based technique, which is highly sensitive and has the potential to detect the small subpopulations of a methylated sequence (133). It is used to detect a sequence corresponding to a particular methylation state (either methylated or unmethylated) in extracted bisulphite modified genomic DNA. Primers were specifically designed to utilize the 53 sequence differences between methylated and unmethylated DNA resulting from sodium bisulphate treatment. For all MSP assays, a series of positive and negative controls plus H20 blank were run simultaneously with each reaction. Reagents: Maxima® Hot Start PCR Master Mix (2X) Primers Bisulfite Converted Universal Methylated Human DNA Standard (Thermo Scientific, Lithuania) (Metabion international, Germany) (ZymoResearch, USA) For a negative methylation control, normal human blood lymphocyte DNA treated with bisulfite was used. Promoter methylation was detected by MSP. Each MSP reaction incorporated approximately 20 ng of bisulfite treated DNA as a template. Sequence primers specific for methylated and unmethylated DNA were obtained from the published data. For AREG and NPTX2, the primer design for these assays was based on bisulfite sequencing results and incorporated 2 CpG dinucleotides that were regularly methylated in glioblastoma and unmethylated in normal adult human brain. All primers are listed in Table 4.8.1. Equipment: MS2 Minishaker (IKA, Wilmington, USA) Centrifuge 5415 R (Eppendorf AG, Germany) Mastercycler epgradient S (Eppendorf AG, Germany) 10/100 μL single-channel pipettes (Eppendorf) Protocol for one sample: Component 2x polymerase mix Primer_M_F or Primer_U_F Primer_M_R or Primer_U_R Water Modified-DNA (20 ng/μL) Volume 10 μL 1 μL 1 μL 6 μL 2 μL Total 20 μL 54 Table 4.8.1. Primers for methylation-specific polymerase chain reaction Primer Tm, °C M_F CGGCGTATATTTTCGGTTTTTATTC 62 M_R GTCTCGATCTCTAAAACAACTCGAT 62 U_F GAGAGTGGTGTATATTTTTGGTTTTTATTT 62 U_R ATCTCAATCTCTAAAACAACTCAAT 62 CASP8 M_F TAGGGGATTCGGAGATTGCGA 58 M_R CGTATATCTACATTCGAAACGA 58 U_F TAGGGGATTTGGAGATTGTGA 58 U_R CCATATATATCTACATTCAAAACAA 58 CD81 M_F CGACGGCGGCGATTTTATCGC 58 M_R GACCTACGAAACGCGAACCG 58 U_F GTGATGGTGGTGATTTTATTGT 58 U_R ACAACCTACAAAACACAAACCAA 58 DcR1 M_F TTACGCGTACGAATTTAGTTAAC 60 M_R TCAACGACCGACCGAAACG 60 U_F GAATTTTTTTATGTGTATGAATTTAGTTAAT 60 U_R CCATCAAACAACCAAAACA 60 DR4 M_F TTCGAATTTCGGGAGCGTAGC 60 M_R GTAATTCAATCCTCCCCGCGA 60 U_F GTAGTGATTTTGAATTTTGGGAGTGTAGT 60 U_R CTCATAATTCAATCCCCACAA 60 GATA4 M_F GTATAGTTTCGTAGTTTGCGTTTAGC 62 M_R AACTCGCGACTCGAATCCCCG 62 U_F TTTGTATAGTTTTGTAGTTTGTGTTTAGT 62 U_R CCCAACTCACAACTCAAATCCCCA 62 GATA6 M_F CGGGGTAGATTTCGGATTCGC 60 M_R CAACCGAACCTCGAACGAACG 60 U_F GTGTGGGGTAGATTTTGGATTTGT 60 U_R AAACAACCAAACCTCAAACAAACA 60 hMLH1 M_F ACGTAGACGTTTTATTAGGGTCGC 60 M_R CCTCATCGTAACTACCCGCG 60 U_F TTTTGATGTAGATGTTTTATTAGGGTTGT 60 U_R ACCACCTCATCATAACTACCCACA 60 NPTX2 M_F CGTGATTTTGTTTTTAATTTCG 60 M_R GAATACAACGATCCCCGA 60 U_F TGTGATTTTGTTTTTAATTTTG 60 U_R CCAAATACAACAATCCCCAA 60 TES M_F TATTGAGTTTGTTTAGTAGGGCGTC 62 M_R AATAACAACCGAACAACTCCG 62 U_F TGAGTTTGTTTAGTAGGGTGTTG 62 U_R ATAACAACCAAACAACTCCAA 62 TFPI2 M_F TTCGTTTCGTATAAAGCGGGTATTC 61 M_R CCGTCAAAAAAAACAACAAAATCG 61 U_F TTTGTTTTGTATAAAGTGGGTATTTGG 61 U_R CATCAAAAAAAACAACAAAATCAAC 61 M, methylated; U, unmethylated; F, forward; R, reverse; Tm, melting temperature. Gene AREG 55 DNA amplification reaction conditions are shown in Table 4.8.2. Table 4.8.2. DNA amplification reaction conditions Step Initial denaturation DNR denaturation Annealing Extension Final extension END Temperature, ºC 95 95 Look table 72 72 4 Duration 5 min 30 s 1 min 1 min 5 min – Cycles – 36-40 – – The amplification products were loaded on a 2% agarose gel with ethidium bromide, and after electrophoresis gels were documented under UV. In a case of both methylated and unmethylated signal appearance in a gel, methylation of the gene was considered. Agarose gel electrophoresis Reagents: TopVision™ Agarose Buffer 10xTBE UltraPure™ 6X DNA Loading Dye O’GeneRuler™ Low Range DNA Ladder, ready-to-use Distillated water Ethidium bromide (Thermo Scientific, Lithuania) (Invitrogen, USA) (Thermo Scientific, Lithuania) (Thermo Scientific, Lithuania) Equipment: Microwave Electronic balance ABJ 120-4M (KERN & Sohn GmbH) Mini horizontal submarine unit HE-33 (Amersham Biosciences) Electrophoresis Power Supply EPS 601 (Amersham Biosciences) Bio-Rad Molecular imager Gel Doc (BIO-RAD laboratories) Protocol: Making the gel 1. Place a casting platform with a well former sideways in a gel stand where you wish to pour the gel. 2. For a 2% gel, add 2 g of high purity, wide range agarose per 100 mL to be made. 3. Add 100 mL of 1X TBE. 56 4. Mix and microwave for about 1-3 minutes. Stop and mix the solution once or twice during the microwaving. 5. Add 0.5 g of ethidium bromide per mL of gel solution from stock solution (10 mg/mL). 6. Pour hot gel into a gel cast on a flat surface avoiding bubbles. 7. When gel solidifies, turn gel and tray to a proper position and fill a gel stand with 1X TBE so that it covers the gel completely. Loading the gel Mix the reaction volumes on a piece of parafilm as follows: 1. First, load a loading dye onto parafilm, add the appropriate volume of the sample, and mix with one pipette. 2. Then, with the same pipette tip, load the sample with the loading pipette into the appropriate well very carefully. Loading buffer (2 μL) was added to each PCR product and 10 μL of this mixture was pipetted into wells of a 2% agarose gel. A DNA ladder was run at the same time to confirm that products were of the expected size. Gels were run for 35-45 minutes at 150 V. DNA was visualized with UV light using a Bio-Rad Molecular imager Gel Doc System. For each primer sequences, samples which were positive displayed visible bands in their corresponding lanes. The MSP assay including the samples of unknown methylation status was only regarded as being successful if there was a visible band in the positive control lanes and no visible bands in both the negative control and H2O blank lane. 4.9. Statistical analysis Statistical analysis was carried out with the software of IBM SPSS Statistics 19 (IBM SPSS Inc., Chicago, IL). Quantitative data are presented as mean and standard deviation (SD). To show the reliability of the estimate, the confidence interval (CI) with 95% confidence level was calculated. To test the statistical hypothesis, the significance level of 0.05 was selected. The Kaplan-Meier method was used to estimate survival functions. For comparing the survival of 2 groups, the log-rank and generalized Wilcoxon tests were used. The Cox proportional hazard regression model was applied to determine the optimal system of independent variables and prognosis-related hazard. The independent sample t test was used to compare the means of 2 groups. The chi-square test was used to test statistical hypothesis about independence of 2 variables. 57 5. RESULTS 5.1. COX7A1, KRT81, AREG, NPTX2, and SPINT1 expression The analysis of gene expression and its regulation is one of the most intriguing fields in cancer research. Promoter methylation affects the expression of many genes in normal and cancer tissues and may play an important role in gliomagenesis. The pharmacologic manipulation of glioma cells with the demethylating agent 5´-aza-dC, combined with genome wide expression profiling, succeeded in unveiling novel candidate genes, which are epigenetically regulated in cancer in general (134) and in gliomas in particular (7, 49). COX7A1, KRT81, AREG, NPTX2, and SPINT1 were among promising candidate genes identified in a previous genome wide study (7). These genes showed robust expression in normal brain samples, their gene promoter harbored a CpG-rich island, and 5`-aza-dC demethylation was accompanied by a significant up-regulation of their expression in glioma cells, suggesting an epigenetic gene regulation (7). One important indication for a potential role of a gene in the development of cancer is its altered expression compared with healthy tissues. To elucidate a potential epigenetic gene regulation of COX7A1, SPINT1, AREG, NPTX2, and KRT81, we investigated their expression in human brain tissues and glioblastoma samples in relation to the methylation status of their gene promoter. RT-PCR is commonly applied to analyze gene expression. By RT-PCR, gene expression can be investigated with high accuracy and excellent reproducibility. RNA sequences, unlike DNA, cannot be multiplied in vitro directly, but the RNA sequence can be converted into cDNA sequences with the assistance of specific enzymes, i.e., reverse transcriptases. In our study, a two-step RT-PCR was used. During the first stage of the experiment, cDNA synthesis was carried out; during the second, cDNA amplification. According to the amplified cDNA content in an electrophoresis gel, the differences in gene expression between glioblastomas and normal brain tissue can be seen. GAPDH (glyceraldehyde 3-phosphate dehydrogenase) amplification was used as an internal control for cDNA quality and quantity. Expression array data for COX7A1, KRT81, AREG, NPTX2, and SPINT1 genes were verified by RT-PCR using cDNA isolated from 22 snap-frozen glioblastoma samples. In addition, we investigated the expression of these genes in cDNA 58 isolated from the normal human brain. Data showed that the housekeeping gene GAPDH was equally expressed in all samples confirming the excellent cDNA quality and the equal cDNA quantity in these samples. Of note, COX7A1, KRT81, AREG, NPTX2, and SPINT1 showed a differential expression pattern in the glioblastoma samples (Figure 5.1.1). GAPDH AREG COX7A1 KRT81 NPTX2 SPINT1 GAPDH AREG COX7A1 KRT81 NPTX2 SPINT1 Figure 5.1.1. Expression of AREG, COX7A1, KRT81, NPTX2, and SPINT1 in glioblastoma samples (1-22) and in normal fetal (FB) and adult human brain (AB) samples The number of glioblastoma and its place in the electrophoresis gel are shown in Table 5.1.1. Gene expression was decreased in some glioblastoma samples. Normal fetal and adult human brain showed robust expression of COX7A1, KRT81, AREG, NPTX2, and SPINT1 suggesting that the down-regulation of these 59 genes in glioblastoma samples was tumor related. As the methylation of these gene promoters might be one of the possible mechanisms of COX7A1, KRT81, AREG, NPTX2, and SPINT1 down-regulation, expression analysis was followed by the analysis of promoter methylation by bisulfite sequencing. Table 5.1.1. The number of glioblastoma and its place in the electrophoresis gel 1. 22164 2. 26882 3. 23218 4. 23480 5. 23316 6. 26718 7. 24040 8. 25290 9. 24672 10. 23480 11. 24528 12. 31118 13. 24702 14. 28590 15. 21636 16. 27824 17. 25108 18. 22334 19. 24440 20. 26494 21. 22584 22. 21660 FB AB 5.2. Bisulfite sequencing of genes promoters DNA sequencing is the method used to determine the primary structure of DNA or order of the nucleotide bases in a molecule of DNA. The bisulfite sequencing method allows a precise analysis of methylation in a certain region by converting all nonmethylated cytosines into thymines, while methylated cytosines remain unchanged. In order to address the possibility of gene regulation by promoter methylation, we decided to perform the bisulfite sequencing of the AREG, COX7A1, KRT81, NPTX2, and SPINT1 gene promoters in selected glioblastoma samples, normal adult human brain specimens, and DNA derived from human leukocytes. 5.2.1. NTPX2 bisulfite sequencing The region of the NPTX2 gene promoter with CpG islands is located on chromosome 7q21.3. The sequence of the promoter region with primer binding sites is shown in Annex. Figure 5.2.1.1 shows the CpG island of the NPTX2 promoter and the fragments of bisulfite sequencing. 60 Figure 5.2.1.1. The CpG island of the NPTX2 promoter and the location of bisulfite sequencing fragments with the number of CpG dinucleotides Three of the NPTX2 fragments were sequenced at the transcription start site. The first fragment is 571 bp in size that covered 53 CpG dinucleotides, the second 184-bp fragment covered 29 CpG dinucleotides, and the third 278-bp fragment covered 40 CpG dinucleotides. Leukocyte, brain, glioblastoma and DNA was sequenced. The results of bisulfite sequencing of the first fragment are shown in Figure 5.2.1.2. The results of bisulfite sequencing of the second fragment are shown in Figure 5.2.1.3. Figure 5.2.1.3. Results of bisulfite sequencing of the second fragment of the NPTX2 promoter in leukocyte and glioblastoma DNA L1 and L2 indicate 2 different samples of leukocytes; 24528, 24702, and 21636, glioblastoma samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 61 Figure 5.2.1.2. Results of bisulfite sequencing of the first fragment of the NPTX2 promoter in leukocyte, brain, and glioblastoma DNA L1-3 indicates 3 different samples of leukocytes; NB1 and NB2, different brain samples; 23316, 24528, 24702, and 21636, glioblastoma samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 62 The results of bisulfite sequencing of the third fragment are shown in Figure 5.2.1.4. Figure 5.2.1.4. Results of bisulfite sequencing of the third fragment of the NPTX2 promoter in leukocyte, brain, and glioblastoma DNA L1-3 indicates 3 different samples of leukocytes; NB1 and NB2, different brain samples; 27824, 24528, 25290, and 21636, glioblastoma samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. The analysis of bisulfite-modified DNA of brain tissue and leukocytes confirmed that the NPTX2 promoter in all 3 fragments had only single 63 scattered methylated CpG dinucleotides. For the analysis of bisulfite sequencing in glioblastoma samples, we chose glioblastoma samples with a different expression profile: samples that showed the expression of cDNA and samples with low cDNA expression. Dense promoter methylation was observed in glioblastoma samples with low cDNA expression. Of note, the NPTX2 gene promoters were unmethylated in glioblastoma samples, which showed high expression of cDNA. These results show an agreement between the promoter methylation and the expression status for this gene. 5.2.2. AREG bisulfite sequencing The AREG CpG island is located at exon 1. The sequence of the promoter region with primer binding sites the location of the exon are shown in Annex. Figure 5.2.2.1 shows the CpG island of the AREG promoter and the fragment of bisulfite sequencing. Figure 5.2.2.1. The CpG island of the AREG promoter and the location of bisulfite sequencing fragment with the number of CpG dinucleotides For AREG, the fragment sequenced at the transcription start site was 412 bp in size and covered 45 CpG dinucleotides. The results are shown in Figure 5.2.2.2. Figure 5.2.2.2. Results of bisulfite sequencing of the AREG CpG islands in brain DNA NB1 and NB2 indicate different brain samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 64 The analysis of bisulfite-modified DNA of brain tissue confirmed that the AREG gene promoter had only single scattered methylated CpG dinucleotides (Figure 5.2.2.2). For the analysis of bisulfite sequencing in glioblastoma samples, we chose glioblastoma samples with a different expression profile: samples that showed the expression of cDNA and samples with low cDNA expression (Figure 5.2.2.3). Dense promoter methylation was observed in glioblastoma samples with low cDNA expression. The AREG promoter was unmethylated in the glioblastoma samples, which showed high expression of cDNA. These results show an agreement between the promoter methylation and the expression status for this gene. Figure 5.2.2.3. Results of bisulfite sequencing of the AREG CpG islands in glioblastoma DNA 25290, 23480, 22164, 28590, and 24060 indicate glioblastoma samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 5.2.3. SPINT1 bisulphite sequencing The region of SPINT1 gene promoter with CpG islands is located on chromosome 15q15.1. The sequence of the promoter region with primer 65 binding sites is shown in Annex. Figure 5.2.3.1 shows the CpG island of the SPINT1 promoter and the fragment of bisulfite sequencing Figure 5.2.3.1. The CpG island of the SPINT1 promoter and the location of bisulfite sequencing fragment with the number of CpG dinucleotides One fragment of the SPINT1 gene promoter was sequenced at the transcription start site. It is a 344-bp fragment that covers 40 CpG dinucleotides. First, 2 brain DNA samples were sequenced. The results of bisulfite sequencing are shown in Figure 5.2.3.2. Figure 5.2.3.2. Results of bisulfite sequencing of the SPINT1 gene promoter in brain DNA NB1 and NB2 indicate different brain samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. The analysis of bisulfite-modified DNA of brain tissue samples revealed that the SPINT1 gene promoter had only single scattered methylated CpG dinucleotides in healthy brain samples. The absence of methylation was associated with the high SPINT1 expression in normal human brain samples. The same analysis with different glioblastoma samples was performed. The results are presented in Figure 5.2.3.3. As can be seen from the results of bisulfite sequencing of glioblastoma samples, the methylation of the SPINT1 gene promoter was not a frequent event. The glioblastoma samples with the high SPINT1 expression did not showed methylation. The glioblastoma samples with the low SPINT1 66 expression accompanied by promoter methylation were observed, but not very frequently. Therefore, our results encourage a more detailed analysis of the entire sequence of the SPINT1 gene promoter in a larger group of glioma samples to clarify the true relationship between promoter methylation and gene expression. Figure 5.2.3.3. Results of bisulfite sequencing of the SPINT1 CpG islands in glioblastoma DNA 23480, 28590, 26882, and 31118 indicate glioblastoma samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 5.2.4. COX7A1 bisulfite sequencing The region of the COX7A1 gene promoter with CpG islands is located on chromosome 19q13.1. The sequence of the promoter region with primer binding sites is shown in Annex. Figure 5.2.4.1 shows the CpG island of the COX7A1 promoter and the fragments of bisulfite sequencing. 67 Figure 5.2.4.1. The CpG island of the COX7A1 promoter and the location of bisulfite sequencing fragments with the number of CpG dinucleotides Two fragments of the COX7A1 gene promoter were sequenced. The first fragment was 309 bp in size that covered 27 CpG dinucleotides; the second fragment was 349 bp in size and harbored 17 CpG dinucleotides. The bisulfite sequencing results of these 2 fragments are shown in Figures 5.2.4.2 and 5.2.4.3. Figure 5.2.4.2. Results of bisulfite sequencing of the first fragment of the COX7A1 promoter in brain DNA NB1 and NB2 indicate different brain samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. Figure 5.2.4.3. Results of bisulfite sequencing of the second fragment of the COX7A1 promoter in brain DNA NB1 and NB2 indicate different brain samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 68 Two fragments of the COX7A1 gene promoter exhibited almost complete methylation of all analyzed sequence clones derived from normal human brain samples. It was decided to investigate the methylation status among different glioblastoma samples. The results of bisulfite sequencing are shown in Figure 5.2.4.4. Figure 5.2.4.4. Results of bisulfite sequencing of the first fragment of the COX7A1 promoter in glioblastoma DNA 22164 and 26882 indicate glioblastoma samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. The results showed that the CpG islands of the COX7A1 gene promoter were abundantly methylated in both glioblastoma samples and normal human brain tissue samples, independent of its gene expression level. 5.2.5. KRT81 bisulphite sequencing The region of the KRT81 gene promoter with CpG islands is located on chromosome 12q13. The sequence of the promoter region with primer binding sites is shown in Annex. Figure 5.2.5.1 shows the CpG island of the KRT81 promoter and the fragments of bisulfite sequencing. Figure 5.2.5.1. The CpG island of the KRT81 promoter and the location of bisulfite sequencing fragments with the number of CpG dinucleotides 69 Two fragments of the KRT81 gene promoter were sequenced at the transcription start site. The first fragment was 324 bp in size and had 26 CpG dinucleotides. The second fragment was 175 bp in size and harbored 11 CpG dinucleotides. Human brain and leukocyte DNA samples were sequenced. The bisulfite sequencing results of both fragments are shown in Figures 5.2.5.2 and 5.2.5.3. Figure 5.2.5.2. Results of bisulfite sequencing of the first fragment of the KRT81 promoter in leukocyte and brain DNA L1 and L2 indicate different samples of leukocytes; NB1 and NB2, different brain samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. Figure 5.2.5.3. Results of bisulfite sequencing of the second fragment of the KRT81 promoter in leukocyte and brain DNA L1 and L2 indicate different samples of leukocytes; NB1 and NB2, different brain samples. The numbers at the top of the figure indicate the numbers of CpG dinucleotides within the fragment. Black, methylated CpG dinucleotide, white, unmethylated. 70 The results showed that the KRT81 gene promoter was frequently methylated in leukocyte and normal human brain samples. Bisulfite sequencing of this gene promoter in glioblastoma samples was not performed as we did not expect that the data could be informative and suitable for future research. These findings suggest that the expression of KRT81 is not regulated by promoter methylation in glioblastomas. Summing up the results of gene expression and sequencing, we can conclude that NPTX2 and AREG were found to be the most promising molecular markers. The data on these genes and their role in gliomagenesis are scarce in the literature. Our data suggest a role of NPTX2 and AREG at least in our subset of glioblastomas. The analysis of bisulfite sequencing showed that glioblastoma samples with low NTPX2 and AREG mRNA expression were highly methylated as compared with normal brain and glioblastoma samples with normal mRNA expression. From these data, it was assumed that mRNA expression was associated with promoter methylation. 5.3. Methylation-specific polymerase chain reaction of AREG and NPTX2 Methylation-specific polymerase chain reaction (MSP) is one of the most commonly used methods for the evaluation of a large number of DNA samples. The methylation frequency of the NPTX2 and AREG gene promoter was evaluated in a series of 22 glioblastoma specimens (Table 5.3.1.). The primer design for these assays was based on the results of bisulfite sequencing and incorporated 2 CpG dinucleotides that were regularly methylated in glioblastoma tumor tissues and unmethylated in the normal adult human brain. The methylation status of the NPTX2 and AREG promoters was evaluated in 22 glioblastomas. NTPX2 promoter methylation was detected in 45.5% (10/22) of the glioblastoma samples, but not in the normal brain specimens. AREG promoter methylation was also detected in 45.5% (10/22) of the glioblastoma samples, but not in the normal brain specimens. It was decided to investigate the methylation of these genes in a larger cohort of patients (n=100) and to find the association between genes methylation, patients’ clinical variables, and overall survival time. 71 Table 5.3.1. Summary of NPTX2 and AREG gene methylation and expression in control brain and glioblastoma samples No. 1 Sample FB AB GBM NPTX2 Methylation 0 0 1 AREG Expression + + + Methylation 0 0 1 2 GBM 1 + 3 GBM 0 + 4 GBM 0 + 5 GBM 1 – 6 GBM 1 – 7 GBM 0 + 8 GBM 0 – 9 GBM 1 – 10 GBM 0 + 11 GBM 1 – 12 GBM 1 + 13 GBM 0 + 14 GBM 1 – 15 GBM 0 + 16 GBM 1 + 17 GBM 1 – 18 GBM 0 + 19 GBM 0 – 20 GBM 0 – 21 GBM 0 – 22 GBM 0 – 1, presence of gene promoter methylation; 0, absence gene was expressed; –, gene was not expressed. Expression + + + 1 + 0 – 0 – 1 – 1 – 0 + 1 – 0 + 0 + 0 + 1 + 1 – 0 + 0 – 1 – 1 – 0 + 0 + 1 – 0 + 0 – of gene promoter methylation; +, 5.4. Characteristics of the study population tested for methylation of 11 genes The data of 100 patients with glioblastoma were analyzed in this study. The median patients’ age at diagnosis was 61.0 years (SD, 1.2; range, 2688). The male-to-female ratio was 1:1.3. The median age of male (n=43) and female (n=57) patients was 58.0 years (SD, 1.8; range, 34-84) and 61.0 years (SD, 1.6; range, 26-88), respectively. The mean survival time of patients was 14.6 months (SD, 16.04; range, 0.20-88.4), while the median survival time was 8.9 months (95% CI, 6.9 to 10.8). More than half (64%) of glioblastoma patients survived less than 12 months after the operation, and the probability to survive longer than 12 and 72 24 months after operation was 36% (95% CI, 26.6%-46.2%) and 21% (95% CI, 13.5%-30.3%), respectively. When the patients were divided into the groups by age (<60 and ≥60 years), Kaplan-Meier survival analysis showed a significant difference between groups with a median survival time of 14.1 months (95% CI, 0.0-29.2) and 4.6 months (95% CI, 4.2-5.0), respectively (P=0.001). The probability to survive more than 12 and 24 months after the operation in the patients aged less than 60 years versus patients aged more 60 years was 58.7% (95% CI, 43.2%-73.0%) and 41.3% (95% CI, 27.0%56.8%) versus 16.7% (95% CI, 7.9%-29.3%) and 3.7% (95% CI, 0.5%12.7%), respectively. The clinical variables were evaluated for a prognostic value separately for age and multifocality using Kaplan-Meier analysis (Figure 5.4.1). The patients younger than 60 years demonstrated much better survival as compared with patients older than 60 years (P<0.0001). The overall survival of the patients with multifocal glioblastomas was worse as compared with the patients with nonmultifocal glioblastomas (P=0.007). Figure 5.4.1. Overall survival of patients with glioblastoma according to age (>60 versus ≤60 years) (top) and tumor multifocality (0, one focus; 1, two and more foci) (bottom) 73 5.5. Gene methylation and its association with clinical variables and survival 5.5.1. AREG methylation Laffaire et al. (6) observed the methylation of the AREG gene promoter in 52% of low-grade gliomas (grade II oligodendrogliomas, grade II astrocytomas). However, there are no data on gene methylation in glioblastomas in the literature so far. To our knowledge, we were the first to show a 58.6% methylation frequency of the AREG gene promoter in glioblastomas (58/99). The representative electrophoresis gel demonstrating the methylation of the AREG gene promoter in glioblastoma specimens is shown in Figure 5.5.1. AREG Figure 5.5.1.1. Methylation status of the AREG CpG island promoter in glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation frequencies of the AREG gene according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.5.1.1. The methylation profile of this gene promoter was significantly associated with 2-year survival (P=0.045). Among 58 AREG-methylated cases, only 8 patients (38.1%) survived longer than 2 years. However, there was no significant difference between the methylation profile and other variables (age, gender, and tumor multifocality), although the association with age was of borderline significance (P=0.065). The methylation of the AREG gene promoter for a prognostic value of overall survival was evaluated using Kaplan-Meier analysis (Figure 5.5.1.2). 74 Table 5.5.1.1. Associations between the methylation of AREG gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 58 (58.6) AREG Unmethylated n (%) 41 (41.4) P N 99 22 (47.8) 36 (67.9) 24 (52.1) 17 (32.1) 0.065 46 53 23 (54.8) 35 (61.4) 19 (45.2) 22 (38.6) 0.541 42 57 53 (57.6) 5 (71.4) 39 (42.4) 2 (28.6) 0.696 92 7 50 (64.1) 8 (38.1) 28 (35.9) 13 (61.9) 0.045 78 21 Figure 5.5.1.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the AREG gene 75 The analysis showed a significant difference in the overall survival between patients with the unmethylated and the methylated AREG gene promoter (P=0.027). The median survival time in the patients with the unmethylated gene was 12.1 months (95% CI, 9.4 to 14.8) versus 6.3 months (95% CI, 4.4 to 8.2) in the patients with the methylated gene. 5.5.2. CASP8 methylation The methylation profile of the CASP8 gene promoter was evaluated in 100 glioblastoma specimens. It was aberrantly methylated in 55% (55/100) of glioblastomas, but not in the control brain specimens. The representative gel electrophoresis demonstrating the methylation of the CASP8 gene promoter in glioblastoma specimens is shown in Figure 5.5.2.1. CASP8 Figure 5.5.2.1. Methylation status of the CASP8 CpG island promoter in glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. Table 5.5.2.1 shows the methylation frequencies of the CASP8 gene according to the patients’ characteristics, tumor multifocality, and survival. The methylation profile of this gene promoter was significantly associated with patients’ age (P=0.044) (Table 5.5.2.1). Among 55 CASP8-methylated cases, 20 patients (43.5%) were younger than 60 years and 35 patients (64.8%) were older than 60 years. However, there was no significant association between the methylation profile and other variables (gender, tumor multifocality, and 2-year survival). The methylation of the CASP8 gene promoter for a prognostic value of overall survival was evaluated using Kaplan-Meier analysis (Figure 5.5.2.2.). 76 Table 5.5.2.1. Associations between the methylation of the CASP8 gene promoter and patients’ age and sex, multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 55 (55.0) CASP8 Unmethylated n (%) 45 (45.0) P N 100 20 (43.5) 35 (64.8) 26 (56.5) 19 (35.2) 0.044 46 54 25 (58.1) 30 (52.6) 18 (41.9) 27 (47.4) 0.686 43 57 51 (54.8) 4 (57.2) 42 (45.2) 3 (42.8) 1 93 7 47 (59.5) 8 (38.1) 32 (40.5) 13 (61.9) 0.090 79 21 Figure 5.5.2.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the CASP8 gene 77 The methylation of this gene promoter was associated with shorter survival (long-rank test, P=0.035). The median survival among patients with the unmethylated CASP8 promoter was 9.9 months (95% CI, 6.4 to 13.4) as compared with 7.7 months (95% CI, 4.8 to 10.6) among their counterparts with the methylated CASP8 promoter. 5.5.3. CD81 methylation Recent glioblastoma methylome studies have shown a CD81 methylation rate of 54% [2, 3]. In this study, the CD81 gene was aberrantly methylated in 48% (48/100) of glioblastomas, but not in the control brain specimens. CD81 Figure 5.5.3.1. Methylation status of the CD81 CpG island promoter in glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation frequencies of the CD81 gene according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.5.3.1. The methylation profile of this gene promoter was not significantly associated with patients’ age and gender, tumor multifocality, and 2-year survival. Kaplan-Meier analysis showed that there was no significant differenece in the overall survival comparing the patients carrying the methylated CD81 gene promoter with those who had the unmethylated CD81 gene promoter (P=0.25) (Figure 5.5.3.2). The median survival time in the patients with the unmethylated gene was 8.9 months (95% CI, 7.0-10.8) versus 8.4 months (95% CI, 4.2-12.5) in patients with the methylated gene. 78 Table 5.5.3.1. Associations between the methylation of the CD81 gene promoter and patients’ age and sex, multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 48 (48.0) CD81 Unmethylated n (%) 52 (52.0) P N 100 22 (47.8) 26 (48.1) 24 (52.2) 28 (51.8) 1 46 54 20 (46.5) 38 (49.1) 23 (53.5) 29 (50.9) 0.842 43 57 45 (48.4) 3 (42.9) 48 (51.6) 4 (57.1) 1 93 7 35 (44.3) 13 (61.9) 44 (55.7) 8 (38.1) 0.219 79 21 Figure 5.5.3.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the CD81 gene 79 5.5.4. DcR1 methylation The methylation profile of the DcR1gene promoter was evaluated in 98 glioblastoma specimens. It was aberrantly methylated in 27.6% (27/98) of glioblastomas, but not in the control brain specimens. The representative electrophoresis gel demonstrating the methylation of the CASP8 gene promoter in glioblastoma specimens is shown in Figure 5.5.4.1. DcR1 Figure 5.5.4.1. Methylation status of the DcR1 CpG island promoter in 14 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB14, glioblastoma samples, NB, normal brain. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation frequencies of the DcR1 gene according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.5.4.1. However, the methylation profile of this gene promoter was not significantly associated with patients’ age and gender, tumor multifocality, and 2-year survival. Kaplan-Meier analysis showed that there was no significant differenece in the overall survival comparing the patients carrying the methylated DCR1 gene promoter with those who had the unmethylated CD81 gene promoter (P=0.55) (Figure 5.5.4.2). The median survival time in the patients with the unmethylated gene was 8.3 months (95% CI, 5.8-10.9) versus 9.7 months (95% CI, 5.8-13.7) in patients with the methylated gene. 80 Table 5.5.4.1. Associations between the methylation of the DcR1 gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 27 (27.6) DcR1 Unmethylated n (%) 71 (72.4) P N 98 12 (27.3) 15 (27.8) 32 (72.7) 39 (72.2) 1 44 54 9 (21.4) 18 (32.1) 33 (78.6) 38 (67.9) 0.263 42 56 27 (29.7) 0 (0) 64 (70.3) 7 (100) 0.185 7 91 22 (27.8) 5 (26.3) 57 (72.2) 14 (73.7) 1 79 19 Figure 5.5.4.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the DcR1 gene 81 5.5.5. DR4 methylation The methylation profile of the DR4 gene promoter was evaluated in 99 glioblastoma specimens. It was aberrantly methylated in 43.4% (43/99) of glioblastomas, but not in the control brain specimens. The representative electrophoresis gel demonstrating the methylation of the DR4 gene promoter in glioblastoma specimens is shown in Figure 5.5.5.1. DR4 Figure 5.5.5.1. Methylation status of the DR4 CpG island promoter in 15 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. Table 5.5.5.1 shows the methylation frequencies of the DR4 gene according to the patients’ characteristics, tumor multifocality, and survival. The methylation profile of this gene promoter was not significantly associated with patients’ age and gender, tumor multifocality, and 2-year survival, although the association between the methylation of the DR4 gene promoter and 2-year survival was of borderline significance (P=0.079) (Table 5.5.5.1). Kaplan-Meier analysis showed that there was no significant differenece in the overall survival comparing the patients carrying the methylated DR4 gene promoter with those who had the unmethylated DR4 gene promoter (P=0.165) (Figure 5.5.5.2). The median survival time in the patients with the unmethylated gene was 9.1 months (95% CI, 6.9-11.3) versus 6.4 months (95% CI, 2.6-10.3) in patients with the methylated gene. 82 Table 5.5.5.1. Associations between the methylation of the DR4 gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 43 (43.4%) DR4 Unmethylated n (%) 56 (56.6%) P N 99 16 (35.6) 27 (50.0) 29 (64.4) 27 (50.0) 0.161 45 54 21 (48.8) 22 (39.3) 22 (51.2) 34 (60.7) 0.415 43 56 41 (44.6) 2 (28.6) 51 (55.4) 5 (71.4) 0.696 92 7 38 (48.1) 5 (25.0) 41 (51.9) 15 (75.0) 0.079 79 21 Figure 5.5.5.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the DR4 gene 83 5.5.6. GATA4 methylation The methylation profile of the GATA4 gene promoter was evaluated in 98 glioblastoma specimens. It was aberrantly methylated in 22.5% (22/98) of glioblastomas, but not in the control brain specimens. The representative electrophoresis gel demonstrating the methylation of the GATA4 gene promoter in glioblastoma specimens is shown in Figure 5.5.6.1. GATA4 Figure 5.5.6.1. Methylation status of GATA4 CpG island promoter in 14 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB14, glioblastoma samples; NB, normal brain. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The associations between the methylation frequencies of the AREG gene and patients’ characteristics, tumor multifocality, and survival were evaluated. The methylation profile of this gene promoter was significantly associated with patients’ age (P=0.027) (Table 5.5.6.1). Among 22 GATA4methylated cases, 5 patients (23%) were younger than 60 years and 17 patients (77%) were older than 60 years. However, there was no significant difference between the methylation profile and other variables (gender, tumor multifocality, and 2-year survival). The Kaplan-Meier analysis showed no significant association between the methylation of the GATA4 gene promoter and overall survival in the patients with glioblastoma (P=0.13) (Figure 5.5.6.2). The median survival time in the patients with the unmethylated gene was 9.0 months (95% CI, 7.5 to 10.6) versus 4.5 months (95% CI, 0.0 to 9.1) in the patients with the methylated gene. 84 Table 5.5.6.1. Associations between the methylation of the GATA4 gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 22 (22.5) GATA4 Unmethylated n (%) 76 (77.5) P N 98 5 (11.4) 17 (31.5) 39 (88.6) 37 (68.5) 0.027 44 54 11 (26.8) 11 (19.3) 30 (73.2) 46 (80.7) 0.464 41 54 20 (22.0) 2 (28.6) 71 (78.0) 5 (71.4) 0.652 91 7 20 (25.6) 2 (10.0) 58 (74.3) 18 (90.0) 0.227 78 20 Figure 5.5.6.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the GATA4 gene 85 5.5.7. GATA6 methylation The methylation profile of the GATA6 gene promoter was evaluated in 100 glioblastoma specimens. It was aberrantly methylated in 68.0% (68/100) of glioblastomas, but not in the control brain specimens. The representative electrophoresis gel demonstrating the methylation of the GATA6 gene promoter in glioblastoma specimens is shown in Figure 5.5.7.1. GATA6 Figure 5.5.7.1. Methylation status of the GATA6 CpG island promoter in 15 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation frequencies of the GATA6 gene according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.5.7.1. None of the variables analyzed was significantly associated the methylation status of this gene promoter. The Kaplan-Meier analysis showed a significant difference in the overall survival between patients with the unmethylated and the methylated GATA6 gene promoter (P=0.025) (Figure 5.5.7.2). The median survival time in the patients with the unmethylated gene was 10.9 months (95% CI, 6.4 to15.6) versus 6.3 months (95% CI, 3.1 to 9.5) in the patients with the methylated gene. 86 Table 5.5.7.1. Associations between the methylation of the GATA6 gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 68 (68.0) GATA6 Unmethylated n (%) 32 (32.0) P N 100 28 (60.9) 40 (74.1) 18 (39.1) 14 (25.9) 0.198 46 54 30 (69.8) 38 (66.7) 13 (30.2) 19 (33.3) 0.830 43 57 62 (66.7) 6 (85.7) 31 (33.3) 1 (14.3) 0.425 93 7 57 (72.2) 11 (52.4) 22 (27.8) 10 (47.6) 0.114 79 21 Figure 5.5.7.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the GATA6 gene 87 5.5.8. hMLH1 methylation Immunohistochemical studies have recently reported a reduced expression of hMLH1 in recurrent compared with primary glioblastomas (53), while hMLH1 gene methylation in gliomas ranges from 0% to 15%. Fukushima et al. (54) showed a higher methylation frequency of the hMLH1 gene in the patients with anaplastic astrocytomas (21.4%, 3/14) than those with GBM (14.8%, 4/27). The methylation profile of the hMLH1 gene promoter was evaluated in 98 glioblastoma specimens. Our study showed only a 2% methylation frequency (2/98), and this is in line with the data of the study by Felsberg et al. (53), who did not observe any hMLH1 methylation in glioblastomas. The representative electrophoresis gel demonstrating the methylation of the hMLH1 gene promoter in glioblastoma specimens is shown in Figure 5.5.8.1. hMLH Figure 5.5.8.1. Methylation status of the hMLH1 CpG island promoter in 15 glioblastoma by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. A low percentage of the methylated cases did not allow us to performed statistical analysis and check for all possible associations. On the other hand, both patients with the methylated hMLH1 gene were 61 years old with much lower as overall median postoperative survival (3.7 and 7.8 months). Fukushima et al. (54) showed an association between hMLH1 and MGMT methylation in the same tumor, and in this study, both patients with the 88 methylated hMLH1 gene had the concomitantly methylated MGMT gene (data not shown). Kaplan-Meier analysis showed no significant difference in the overall survival between patients with the unmethylated and the methylated GATA6 gene promoter (P=0.236) (Figure 5.5.8.2). The median survival time in the patients with the unmethylated gene was 8.9 months (95% CI, 6.9 to 10.9) versus 3.7 months (95% CI, not applicable) in the patients with the methylated gene. Figure 5.5.8.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the hMLH1 gene 5.5.9. NTPX2 methylation The data on NPTX2 epigenetic silencing in glioblastoma are scarce. In our study, the methylation profile of the DR4 gene promoter was evaluated in 98 glioblastoma specimens. To our knowledge, we were the first to show a 53.1% methylation frequency of the NPTX2 gene promoter (52/98) in patients with glioblastoma. The representative electrophoresis gel demonstrating the methylation of the DR4 gene promoter in glioblastoma specimens is shown in Figure 5.5.9.1. 89 NTPX2 Figure 5.5.9.1. Methylation status of the NTPX2 CpG island promoter in 15 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation profile of this gene promoter was significantly associated with tumor multifocality (P=0.049) (Table 5.5.9.1). Table 5.5.9.1. Associations between the methylation of the NTPX2 gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Methylated n (%) 52 (53.1) NTPX2 Unmethylated n (%) 46 (46.9) P N 98 26 (56.5) 20 (43.5) 0.549 46 26 (50.0) 26 (50.0) 24 (58.5) 28 (49.1) 17 (41.5) 29 (50.9) 0.415 41 57 51 (56.0) 1 (14.3) 40 (44.0) 6 (85.7) 0.049 91 7 42 (54.5) 10 (47.6) 35 (45.5) 11 (52.4) 0.627 77 21 52 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Among 52 NTPX2-methylated cases, only 1 patient (14.3%) had a multifocal glioblastoma as compared with 51 patients (56.0%) with without multifocal glioblastomas. No significant associations between the 90 methylation status of the NTPX2 gene promoter and age, gender, and survival were documented. Kaplan-Meier analysis showed no significant difference in the overall survival between patients with the unmethylated and the methylated NPTX2 gene promoter (P=0.554) (Figure 5.5.9.2). The median survival time in the patients with the unmethylated gene was 8.3 months (95% CI, 5.4 to 11.2) versus 9.7 months (95% CI, 5.8 to 13.6) in the patients with the methylated gene. Figure 5.5.9.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the NPTX2 gene The methylation of the NPTX2 gene promoter was a frequent event in our series of glioblastomas, but the importance of the gene in glioblastoma remains to be elucidated. 5.5.10. TES methylation Some studies showed that TES was more frequently methylated in glioblastomas (58%-69%) than low-grade gliomas (28%-30%) (2, 6, 7). In agreement to the previously published studies, we observed a 66.7% methylation frequency of the TES gene promoter in 99 glioblastoma specimens. The representative electrophoresis gel demonstrating the 91 methylation of the DR4 gene promoter in glioblastoma specimens is shown in Figure 5.5.10.1. TES Figure 5.5.10.1. Methylation status of the TES CpG island promoter in 15 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation frequencies of the TES gene according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.5.10.1. Table 5.5.10.1. Associations between the methylation of the TES gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 66 (66.7) TES Unmethylated n (%) 33 (33.3) P N 99 29 (63.0) 37 (69.8) 17 (37.0) 16 (30.2) 0.526 46 53 27 (64.3) 39 (68.4) 15 (35.7) 18 (31.6) 0.673 42 57 61 (66.3) 5 (71.4) 31 (33.7) 2 (28.6) 1 92 7 53 (67.9) 13 (61.9) 25 (32.1) 8 (38.1) 0.611 78 21 92 None of the variables analyzed was significantly associated the methylation status of this gene promoter (P>0.05). Kaplan-Meier analysis showed no significant difference in the overall survival between patients with the unmethylated and the methylated TES gene promoter (P=0.947) (Figure 5.5.10.2). The median survival time in the patients with the unmethylated gene was 8.9 months (95% CI, 5.8 to 12.1) versus 8.4 months (95% CI, 5.9 to 10.8) in the patients with the methylated gene. Figure 5.5.10.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the TES gene TES methylation needs further analysis to clarify the meaning of such a high level of epigenetic down-regulation in glioblastoma. 5.5.11. TFPI2 methylation The methylation profile of the TFPI2 gene promoter was evaluated in 99 glioblastoma specimens. It was aberrantly methylated in 22.2% (22/99) of glioblastomas, but not in the control brain specimens. The representative electrophoresis gel demonstrating the methylation of the DR4 gene promoter in glioblastoma specimens is shown in Figure 5.5.11.1. 93 TFPI2 Figure 5.5.11.1. Methylation status of the TFPI2 CpG island promoter in 14 glioblastoma specimens by the methylation-specific PCR assay Molecular weight markers are shown on the left. mDNA indicates methylated DNA control; uDNA, unmethylated DNA control; W, water control; GB1-GB15, glioblastoma samples; NB, normal brain. The presence of visible PCR products in the lanes marked “U” indicates the presence of unmethylated genes and “M” indicates the presence of methylated genes. The methylation frequencies of the TFPI2 gene according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.5.11.1. Table 5.5.11.1. Associations between the methylation of the TFPI2 gene promoter and patients’ age and sex, tumor multifocality, and 2-year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 Methylated n (%) 22 (22.2) TFPI2 Unmethylated n (%) 77 (77.8) P N 99 6 (13.3) 16 (29.6) 39 (86.7) 38 (70.4) 0.057 45 54 11 (26.2) 11 (19.3) 31 (73.8) 46 (80.7) 0.468 42 57 21 (22.8) 1 (14.3) 71 (77.2) 6 (85.7) 1 92 7 21 (26.9) 1 (4.8) 57 (73.1) 20 (95.2) 0.037 78 21 The methylation profile of this gene promoter was significantly associated with survival (P=0.037) (Table 5.5.11.1). Among 22 TFPI294 methylated cases, only 1 patient (4.8%) survived longer than 2 years. No significant associations between the methylation status of the TFPI2 gene promoter and age, gender, and tumor multifocality were documented, although the association with age was of borderline significance (P=0.057). These results suggest that the CpG island methylation in the TFPI2 gene may be an important prognostic factor for glioblastoma. To test this hypothesis, we evaluated overall survival using Kaplan-Meier analysis. The analysis showed a significant difference in the overall survival between patients with the unmethylated and the methylated TFPI2 gene promoter (P=0.047) (Figure 5.5.11.2). The median survival time in the patients with the unmethylated gene was 9.6 months (95% CI, 7.3 to 11.9) versus 6.1 months (95% CI, 2.9 to 9.4) in the patients with the methylated gene. Figure 5.5.11.2. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the methylation status of the TFPI2 gene 95 5.6. Gene comethylation and its associations with survival Further we aimed to evaluate the concomitant methylation of 11 genes (AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, TES, and TFPI2). A total of 100 glioblastoma samples were ranked in an ascending order by the number of the methylated genes in them. All results are summarized in Figure 5.6.1. (The smallest number of the methylated genes at the top; and the greatest, at the bottom). The methylation analysis of 11 genes in 100 glioblastomas showed that the most frequently methylated genes were GATA6 (68%) and TES (66.7%), while the least frequently methylated gene was hMLH1 (2%). The overwhelming majority (98%) of the glioblastomas were methylated in at least 1 of the 11 genes tested. Only 2% of the glioblastomas had no detectable gene methylation, and 1% had the concomitant methylation of the 10 genes tested. More than half (52%) of the glioblastoma samples had the comethylation of 4-6 genes. Moreover, the percentage distribution of glioblastoma samples by the number of methylated genes in each specimen was evaluated. The results are depicted in Figure 5.6.2. As can be seen from the figure, the methylation of 5 and 6 genes was the most common in the glioblastoma samples (20% in both cases). Figure 5.6.2. The percentage distribution of glioblastoma specimens by the number of methylated genes in each glioblastoma sample 96 Figure 5.6.1. Analysis of the methylation profile of 11 genes in 100 glioblastoma samples The numbers on the right side indicate the number of methylated genes in samples. The percentages on the bottom indicate the methylation frequency of a particular gene. Reddish cells indicate the methylated gene; greenish, unmethylated gene; and without color, no data about the methylation of a particular gene. 97 Next, we aimed to determine the pairs of comethylated genes, as it is known that the functions of some genes investigated in this study are interrelated in the cell. In our series of glioblastomas, the following genes were comethylated: AREG and TES (chi-square test, P=0.021), AREG and GATA6 (chi-square test, P=0.001), AREG and NPTX2 (chi-square test, P=0.007), TES and GATA6 (chi-square test, P=0.049), NPTX2 and TES (chisquare test, P=0.006), TFPI2 and GATA4 (chi-square test, P=0.02), TFPI2 and DR4 (chi-square test, P=0.01), and DR4 and GATA6 (chi-square test, P=0.034). Some pairs of the comethylated genes showed comethylation associations of borderline significance: CASP8 and DcR1 (chi-square test, P=0.079), CASP8 and DR4 (chi-square test, P=0.095), AREG and DR4 (chisquare test, P=0.059), TES and DR4 (chi-square test, P=0.075), CD81 and GATA4 (chi-square test, P=0.076), and NTPX2 and GATA6 (chi-square test, P=0.088). Further, we aimed to determine the median survival time of the patients with glioblastoma by the number of the comethylated genes. The data of a total of 90 patients were included in this analysis as the data on the methylation of 1 or more genes were missing in 10 patients. Table 5.6.1 shows that the patients can be divided into 2 groups according to the medial survival time: patients having 0-4 comethylated genes (0-4 group) and those with 5-11 comethylated genes (5-11 group). Table 5.6.1. Median patients’ survival time according to the number of comethylated genes No. of comethylated Median survival SE genes time, months 0 8.9 NA 1 7.6 1.7 2 12.9 5.8 3 8.9 1.1 4 10.8 4.5 5 4.5 0.2 6 9.7 2.0 7 4.1 3.7 8 4.6 2.2 9 20.7 NA 10 7.7 NA Overall 8.3 0.8 NA, not applicable as there was only the one patient. 98 95% confidence interval NA 4.3-10.9 1.6-24.3 6.6-11.1 1.9-19.7 4.1-4.8 5.7-13.6 0.0-11.3 0.2-9.0 NA NA 6.8-9.8 There were 39 patients (43.3%) in the 0-4 group and 51 patients (56.7%) in the 5-11 group. The comethylation frequencies in the 0-4 and 5-11 groups according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.6.2. No significant associations between the comethylation status and patients’ age and gender, tumor multifocality, and survival were documented, although the associations between the comethylation status and patients’ age as well as 2-year survival were of borderline significance (P=0.090 and P=0.060, respectively) Table 5.6.2. Associations between the number of the comethylated genes (04 versus 5-11 genes) and patients’ age and sex, tumor multifocality, and 2year survival Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 SUM11 0-4 5-11 comethylated comethylated genes genes n (%) n (%) 39 (43.3) 51 (56.7) P N 90 21 (53.8) 18 (35.3) 18 (46.2) 33 (64.7) 0.090 39 51 16 (45.7) 23 (41.8) 19 (54.3) 32 (58.2) 0.828 35 55 35 (42.2) 4 (57.1) 48 (57.8) 3 (42.9) 0.461 83 7 28 (38.5) 11 (64.7) 45 (61.6) 6 (35.3) 0.060 73 17 Kaplan-Meier analysis showed that there was no significant difference in the overall survival comparing the 0-4 group with the 5-11 group (P=0.094) (Figure 5.6.3). The median survival time in the 0-4 group was 9.6 months (95% CI, 5.9-13.3) versus 6.3 months (95% CI, 3.1-9.5) in the 5-11 group. Moreover, Cox proportional hazards analysis reveled that SUM11 did not have any prognostic value (HR, 1.42; 95% CI, 0.91-2.35; P=0.12). Considering the data obtained, it was assumed that the methylation of not all genes could have an impact on overall survival after surgery. 99 Figure 5.6.3. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the comethylation groups (0-4 and 5-11) Further, it was aimed to identify the combination of genes that could help prognosticate patients’ survival after surgery and could be used in diagnostics in clinical practice. Therefore, to analyze further the impact of the methylation status of the individual gene promoters, a univariate analysis with the use of the Cox proportional hazards model was performed. Table 5.6.3. Results of analysis with the Cox proportional hazards model Gene DcR1 TFPI2 GATA4 CASP8 CD81 DR4 GATA6 AREG hMLH1 NTPX2 TES HR (95% CI) P value 0.86 (0.53-1.40) 1.67 (1.00-2.77) 1.47 (0.89-2.44) 1.59 (1.03-2.47) 0.78 (0.51-1.19) 1.35 (0.88-2.07) 1.69 (1.06-2.69) 1.63 (1.05-2.51) 2.29 (0.56-9.49) 1.14 (0.74-1.75) 0.99 (0.63-1.54) 0.55 0.05 0.13 0.04 0.25 0.17 0.03 0.03 0.25 0.56 0.95 100 The analysis reveled that the methylation of the following gene promoters were significant in predicting the overall survival: TFPI2 (HR=1.67; 95% CI, 1.00-2.77; P=0.05), CASP8 (HR=1.59; 95% CI, 1.032.47; P=0.04), GATA6 (HR=1.69; 95% CI, 1.06-2.69; P=0.03) and AREG (HR=1.63; 95% CI, 1.05-2.51; P=0.03) (Table 5.6.3). These 4 genes and 2 additional genes (GATA4 and DR4; hazard ratios P<0.2) were combined in a set of 6 genes (SUM6) and were analyzed more detailed. Cox proportional hazards analysis showed that SUM6 cases were more likely to have a shorter survival (HR, 2.2; 95% CI, 1.32-3.68; P=0.003) indicating that this set of the genes could be a prognostic marker to identify the outcome of patients with glioblastoma. The overall survival in patients with glioblastoma according to the comethylation of these 6 genes is depicted in Figure 5.6.4. Kaplan-Meier analysis showed a significant difference in the overall survival comparing the groups with the different number of the comethylated genes (P=0.013). Figure 5.6.4. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the number of the comethylated genes (AREG, CASP8, DR4, GATA4, GATA6, and TFPI2) 101 Further, the median survival time of the patients with glioblastoma by the number of the comethylated genes in a set of 6 genes (SUM6) was calculated. Table 5.6.4 shows that the patients can be divided into 2 groups according to the medial survival time: patients having no methylated genes or only 1 methylated gene (0-1 group) and those with 2-6 comethylated genes (2-6 group). Table 5.6.4. Median patients’ survival time according to the number of comethylated genes in a set of 6 genes ((AREG, CASP8, DR4, GATA4, GATA6, and TFPI2) No. of comethylated genes 0 1 2 3 4 5 6 Overall Median survival time, months 14.1 15.7 9.1 6.2 6.4 3.7 7.6 8.9 SE 4.3 3.4 1.1 3.9 2.1 3 5.8 0.9 95% Confidence Interval 5.6-22.6 9.1- 22.3 7.0-11.1 0.4- 12.1 2.4-10.5 0-9.6 0-18.9 7.1-10.7 The comethylation frequencies in the 0-1 and 2-6 groups according to the patients’ characteristics, tumor multifocality, and survival are shown in Table 5.6.5. Table 5.6.5. Associations between the number of the gene methylated (0-1 versus 2-6 genes) and patients’ age and sex, tumor multifocality, and 2-year survival SUM6 Variable Overall Age, years <60 ≥60 Gender Male Female Multifocal No Yes Survival, months <24 ≥24 0-1 genes methylated n (%) 25 (26.3) 2-6 genes methylated n (%) 70 (73.7) 16 (38.1) 9 (17.0) 26 (61.9) 44 (83.0) 0.030 42 53 8 (20.5) 17 (30.5) 31 (79.5) 39 (69.6) 0.350 39 56 24 (27.3) 1 (14.3) 64 (72.3) 6 (85.7) 0.670 88 7 14 (18.4) 11 (57.9) 62 (81.6) 8 (42.1) 0.001 76 19 102 P N 95 There were 25 patients (26.3%) in the 0-1 group and 70 patients (73.7%) in the 2-6 group. There were significant associations between the comethylation status and patients’ age as well as survival (P=0.030 and P=0.001, respectively). Kaplan-Meier analysis showed that a significant difference in the overall survival comparing the 0-1 group with the 2-3 group (P=0.002) (Figure 5.6.5). The median survival time in the 0-1 group was 15.7 months (95% CI, 10.5-20.9) versus 6.4 months (95% CI, 4.2-8.7) in the 2-6 group. Figure 5.6.5. Kaplan-Meier estimation of overall survival in patients with glioblastoma according to the comethylation groups (0-1 and 2-6) 103 DISCUSSION Microarray analysis coupled with the pharmacologic demethylation of cultured tumor cells is a useful tool for identifying the genome-wide epigenetic events and has been successfully implemented for this purpose in other cancer types in the previous studies (134, 135). However, genes identified by this method have to be necessarily examined more detailed to validate a potential epigenetic gene regulation and to elucidate their importance in cancer development. Multiple independent techniques should be implemented to validate these candidate genes. In our study, the expression of AREG, COX7A1, KRT81, NPTX2, and SPINT1 were investigated first in normal human brain specimens. Normal fetal and adult human brain samples showed robust expression of AREG, COX7A1, KRT81, NPTX2, and SPINT1 suggesting that the down-regulation of these genes in glioblastoma samples was tumor related. The up-regulation of their expression following 5´-aza-dC-mediated DNA demethylation suggested promoter methylation as a mechanism for gene silencing (7). Surprisingly partial bisulfite sequencing of the COX7A1 and KRT81 promoters revealed abundant promoter methylation both in normal human brain and glioblastoma samples, independent of their individual expression levels. Our results do not support the classical hypothesis that promoter hypermethylation leads to gene silencing. They might suggest that the expression of these genes might follow other mechanisms of regulation than promoter methylation in gliomas. However, these results might also be explained by other hypotheses. Some reports have shown that histone deacetylase inhibitors lead to gene reactivation from hypermethylated promoters without any changes in DNA methylation at the promoter level or that the inhibition of SIRT1 reactivates silenced cancer genes without the loss of promoter DNA hypermethylation (136, 137). Another hypothesis suggests that there are some transcription factors, i.e. SP1 or NRF1, that are capable to bind to methylated DNA. COX7A1 and KRT81 transcription might be regulated by one of these transcription factors, explaining their expression despite methylated promoter sequences (138, 139). The third hypothesis relates to the bisulfite sequencing method itself. Bisulfite sequencing cannot discriminate 5mC from 5hmC. The recent discovery that 3 members of the TET protein family can convert 5mC into 5hmC has provided a potential mechanism leading to DNA demethylation (8, 44, 46). We cannot rule out the possibility that in our study not 5mC but rather 5hmC was detected. All these hypotheses have to be addressed in future investigations in order to elucidate the functional contribution of these genes to gliomagenesis. 104 The glioblastoma samples with the low SPINT1 expression accompanied by promoter methylation were observed, but not very frequently. Therefore, our results encourage a more detailed analysis of the entire sequence of the SPINT1 gene promoter in a larger cohort of glioma samples to clarify the true relationship between promoter methylation and gene expression. Summing up the results of gene expression and sequencing, we can conclude that NPTX2 and AREG were found to be the most promising molecular markers. There is no more information about these genes and their role in gliomagenesis. The findings of this study suggest a role of NPTX2 and AREG at least in our subset of glioblastomas. The analysis of bisulfite sequencing showed that glioblastoma samples with low NTPX2 and AREG mRNA expression were highly methylated as compared with normal brain and glioblastoma samples with normal mRNA expression. From these data, it was assumed that mRNA expression was associated with promoter methylation. However, the prognostic value of these biomarkers could not be ascertained, and larger-scale prospective studies are warranted. Using a MSP reaction, primary glioblastomas were analyzed for the methylation of 11 gene promoters and its association with patients’ survival. AREG is a member of the EGF family and a ligand for EGFR (140). Amphiregulin is known as a bifunctional growth-modulating glycoprotein acting stimulatory to several human fibroblast cell lines, while being inhibitory to some neuroblastoma and adenocarcinoma cell lines (141). AREG can either stimulate or inhibit the growth of lung cancer cells, and this is dependent on a biological setting (56). Various studies have highlighted the functional role of AREG in several aspects of tumorigenesis, including self-sufficiency in generating growth signals, limitless replicative potential, tissue invasion and metastasis, angiogenesis, and resistance to apoptosis (57). AREG is associated with the induction of genes involved in invasion and migration, such as cytokines and matrix metalloproteases (142). It is overexpressed in various types of human cancer: colon, breast, pancreas, and lung cancer, and recently, the lack of AREG expression in GBM cell lines has been shown (142-146). There is a growing body of data on AREG promoter methylation in various tumors: gastric, high-grade serous carcinomas, and human bladder tumor cell line T24 (58-60). Microarray analysis coupled with the pharmacologic demethylation of glioblastoma cells showed that AREG is one of the novel tumor suppressor candidate genes (7). According to Laffaire et al., AREG methylation was observed in 52% of low-grade gliomas (grade II oligodendrogliomas, grade II astrocytomas) (6). However, there are no data on gene methylation in glioblastomas in the 105 literature so far. To our knowledge, we were the first to show a 58.6% methylation frequency of the AREG gene promoter in glioblastomas (58/99), and gene methylation was significantly associated with older patient age and shorter postoperative survival. Unmethylated AREG was associated with significantly prolonged survival as compared with that of patients with methylated gene. To our knowledge, we were the first to show an association between AREG gene methylation and poor patients’ outcome in glioblastoma. Interestingly, the comethylation of AREG with TES and NPTX2 was a frequent event in glioblastomas; all the genes are functionally related to regulation of the invasive phenotype of the cell. The hMLH1 gene is one of the MMR genes. The inactivation of genes involved in MMR is associated with high microsatellite instability in various cancers, albeit in sporadic gliomas, typical microsatellite instability is rare (102, 147). However, the main mechanism in sporadic cases of colorectal, endometrial, and gastric cancers with microsatellite instability is transcriptional hMLH1 gene inactivation through its promoter methylation (148). Methylation at the CpG islands in the promoter sequence and the resulting loss of expression of the hMLH1 gene were reported in colon (102, 149), gastric cancer (150), and brain gliomas (54, 151). The promoter methylation of MMR genes plays a causal role in the loss of mismatch repair functions that modulate cytotoxic pathways in response to DNA damaging agents (54). Associations between MMR mutations and MGMT methylation, and resistance to alkylating agent temozolomide that is the current standard of care for glioblastoma patients have been suggested (152). Immunohistochemical findings recently reported on reduced expression of hMLH1 in recurrent compared to primary glioblastomas (53). The methylation frequency of the hMLH1 gene in gliomas ranges from 0% to 15% (53, 54). Fukushima et al. showed a higher methylation frequency of hMLH1 in anaplastic astrocytomas (21.4%, 3/14) as compared with glioblastoma multiforme (14.8%, 4/27) (54). In line with a study by Felsberg et al., who did not observe any methylation of hMLH1 in glioblastomas, our study showed only a 2.0% methylation level in glioblastomas (2/98) (53). Both patients with the methylated hMLH1 gene were 61 years old with much lower as overall median postoperative survival (3.7 and 7.8 months). Fukushima et al. (54) showed an association between hMLH1 and MGMT methylation in the same tumor, and in this study, both patients with the methylated hMLH1 gene had the concomitantly methylated MGMT gene (data not shown). However, there were no significant relationships between hMLH1 methylation and patients’ survival in our glioblastoma samples. 106 The NPTX2 gene encodes a member of the family of neuronal pentraxins, synaptic proteins that are related to C-reactive protein, and has a wide distribution in various tissues and is normally expressed in the CNS (153). In vitro data with pancreatic cancer cell lines showed that NPTX2 suppressed tumor cell growth, invasion, and migration and affected apoptosis (112). Carlson et al. showed increased NPTX2 expression as an independent negative predictor of survival with the highest levels of edema in glioblastoma (154). There is a lack of information on NPTX2 methylation silencing in glioblastoma. For the first time, a NPTX2 methylation frequency of 53% was determined in glioblastomas, but no significant associations between the gene methylation status and patients’ age and gender as well as survival were found. Recently, the TES gene has been identified as highly methylated in glioblastomas (2, 6, 7). It was confirmed by the tumor-related methylation of TES in cultured glioblastomas and glioblastoma cell lines (7). When the TES gene becomes methylated, the cells loose internetworks, and this allows them to migrate and tumor infiltrates brain. Gunduz et al. showed an association between TES down-regulation and worse patients’ outcome in head and neck squamous cell carcinomas (155). The down-regulation of TES was shown to be correlated with tumor differentiation and prognosis in gastric cancer (156), and it was densely methylated in acute lymphoblastic leukemia (121). It has been shown that TES appears to be more frequently methylated in glioblastoma (58%-69%) as compared to low-grade glioma (28%-30%) (2, 6, 7). In agreement to the previous studies, our study showed a similar TES methylation frequency (66.7%) in glioblastoma. Moreover, TES and a few other genes were found to be comethylated. However, there were no significant associations between the methylation status of TES and patients’ age and gender, tumor multifocality, and survival in this study. The methylation of the TES gene promoter needs further analysis to clarify the meaning of such a high level of epigenetic down-regulation in glioblastoma pathogenesis. The transcription factor GATA6 with a methylation frequency of 68% was one of the most frequently methylated genes in our study. GATA6 is one of 6 members of the mammalian GATA family of transcription factors that direct cell proliferation, differentiation, and inhibits apoptosis (157). Recently, it has been identified as a tumor suppressor gene in the CNS tumors with a gene expression loss of 90% and promoter methylation of 30%-48% in glioblastoma (2, 50-52). It was shown that the loss of GATA6 results in enhanced astrocyte proliferation and transformation (52). Reduced GATA6 expression was observed in colon and ovarian carcinomas (92, 158). 107 In our series, the methylation status of the GATA6 gene promoter was not associated with patients’ age and sex or tumor multifocality. Kaplan-Meier analysis showed a significant association between the methylation of GATA6 and overall patients’ survival. In agreement with the finding of the methylome study by Martinez et al. (2), where the importance of GATA6 methylation in patients’ survival has been shown for the first time, our results suggest that the methylation of GATA6 is a frequent event and is highly important for survival of patients with glioblastoma. The CD81 gene promoter was methylated in 48% of the glioblastomas in our study. This gene is a member of the membrane-embedded tetraspanin superfamily, which was found to be silenced by methylation in multiple myeloma cell lines (68). CD81 participates in several functions such as cell adhesion and signal transduction (159). It was shown that the methylation frequency of CD81 in glioblastomas was 54% (2, 6). Our study showed no significant associations between the methylation status of this gene and patients’ age and gender, tumor multifocality, and survival. The analysis of the methylation status of proapoptotic genes in glioblastomas revealed that CASP8 and DR4 were methylated in 55% and 48% of the samples, respectively. DR4 inactivation by promoter methylation has been previously reported in osteosarcomas (160), melanomas (161), medulloblastomas (162), gastric carcinomas (6) and glioblastomas (2, 26, 49, 50). The DR4 methylation frequency in glioblastomas varied from 25% to 70% in different studies. We hypothesize that this variation could be attributed to the heterogeneity of glioblastomas. Our study showed no significant associations between the methylation status of this gene and patients’ age and gender, tumor multifocality, and survival.. In contrast to the findings of Elias et al. (49) and Hervouet et al. (50), who showed a 10%-30% methylation level of CASP8 in glioblastoma, more than half of tumors were methylated (55%) in our sample. In addition, we noted a slight tendency for a relationship between the methylation of 2 TRAIL associated proapoptotic genes CASP8 and DR4 in glioblastoma: both genes were comethylated in 28.4% (21/74) of the tumors. The latter finding generally is in line with the findings of Elias et al. (49), who found a 10% DR4 and CASP8 co-methylation level in GBM. It is known that DR4 and CASP8 are factors affecting nonmitochondrial apoptotic pathway, and the DR4 loss of expression, which was shown to be mediated by promoter methylation, attenuates apoptosis and is associated with glioma cell resistance to the pro-apoptotic ligand therapy (known as TRAIL resistance) (49). The importance of CASP8 in TRAIL resistance in gliomas has been reported as well (63), while other studies showed CASP8-independent and DR4-specific TRAIL sensitivity (49). 108 When looking for the associations between the methylation status and clinical variables, it should be mentioned that CASP8 gene methylation was associated with patients’ age. Glioblastoma patients with methylated CASP8 were significantly older than those with unmethylated CASP8. As age is known to play a significant role in patients’ survival, the methylation status of CASP8 could be of an additive value in tumor genesis and patients’ outcome. Our data showed that the methylation status of GATA6 and CASP8, but not CD81 and DR4 were independent factors for survival of patient with glioblastoma. Similar to the study by Harvouet et al. (50), the associations between CASP8 gene methylation and survival showed in our study gene methylation being important factor in worse outcome of patients. The patients with the unmethylated CASP8 gene, as a favorable genotype, mostly had prolonged survival as compared with the patients having tumors with the methylated CASP8 gene. Our data complement the findings of Martinez et al. (26) on the significance of CASP8 methylation in glioblastoma progression. Cox regression analysis revealed CASP8 and GATA6 as the independent predictors of survival in patients with glioblastoma. Patients with methylated CASP8 and GATA6 were more likely to have poor survival (HR=1.59; 95% CI, 1.03-2.47; P=0.04; and HR=1.69; 95% CI, 1.06-2.69; P=0.03). Contrary to our results, Cecener et al. (51) and Hervouet et al. (50) reported no associations between the methylation status of GATA6 and CASP8 and survival in glioblastoma patients. The loss of GATA4 expression due to promoter hypermethylation has been reported in primary colorectal, gastric, esophageal, lung, and ovarian cancer (163, 164). To analyze the potential of GATA4 as a methylation marker in glioblastoma, we analyzed a large series of glioblastoma samples and showed that in glioblastoma, GATA4 was not so abundantly methylated (23.2%) when compared with colorectal cancer (70%) (163) or sporadic gastric carcinomas (53.8%) (164). The loss of GATA4 protein expression was observed in about 57.7% of GBM samples (94). In our study, GATA4 methylation was detected only in 23.2% of cases, and this may suggest that the GATA4 expression is regulated by different mechanisms or mutations. Probably for this reason, GATA4 promoter methylation was not found to be significantly associated with survival in comparison with the results obtained in a study by Agnihotri et al. (94). The members of the tumor necrosis factor receptor or death receptor family were found to be frequently methylated in glioblastoma: TNFRSF10D promoter was methylated in 100% (6) and TNFRSF10A in 68% (2) of glioblastomas. The methylation of the DcR1 gene promoter was 109 determined in 21% of low-grade gliomas (WHO grade II) (6). The data on DcR1 promoter methylation in glioblastoma are scarce. A study by Martinez et al. involving 16 glioblastoma patients did not reported the methylation of the DcR1 promoter (26). We have assumed that this percentage should be much higher in glioblastomas. Our study found that DcR1 was methylated in 27.6% of glioblastoma samples, but not in the normal brain specimens, which implies that the epigenetic silencing of the DcR1 gene promoter might be involved in gliomagenesis. This percentage is close to a 37% methylation level in lung cancer cell lines or 30.7% in ovarian tumors and differs from 78.0% in prostate cancer (83, 165, 166). These findings suggest that the prognostic value of DcR1promoter methylation could be tissue specific. Although the DcR1 gene promoter was found to be methylated in our series of glioblastomas, the methylation profile was not associated with patients’ sex and age, tumor multifocality, or survival. The ability of glioblastoma cells to infiltrate the surrounding brain parenchyma critically limits the effectiveness of current treatments (167).Tumor invasion is a complex, multistep process, and the mechanisms resulting in the degradation of the ECM and tumor cell migration and invasion have not been completely understood yet (129). The matrix degradation can be promoted by the imbalance between proteolytic enzymes (proteases) and their inhibitors (168, 169). Additional studies have shown that the tissue factor pathway inhibitor 2 inhibits tumor-related angiogenesis and some members present in the ECM therefore implicates tumor invasion and progression (170). It was shown previously that TFPI-2, a broad range proteinase inhibitor, is highly expressed in low-grade gliomas, but, minimally expressed or undetectable in glioblastomas, and that the enforced expression of this gene reduces the invasive properties of brain tumor cells (171). The aberrant methylation of TFPI-2 promoter CpG islands in human cancers and cancer cell lines was widely documented to be responsible for the diminished expression of mRNA encoding TFPI-2 and decreased or inhibited synthesis of TFPI-2 protein during cancer progression (172). The transcriptional silencing of TFPI-2 by hypermethylation in the promoter region has recently been demonstrated in many kinds of human cancers: TFPI-2 promoter was methylated in 88.6% (62/70) of nasopharyngeal carcinoma primary tumors (129), 93% (51/55) of colorectal tumor samples (173), 27.1% (36/133) of non–small cell lung cancer (174), and 18% (7/38) of primary gastric carcinomas (173, 175). The aberrant methylation of TFPI-2 was also detected in 73% (102/140) of pancreatic 110 cancer xenografts and primary pancreatic adenocarcinomas and was more likely in older patients with pancreatic cancer (176). However, to our knowledge, the methylation status of the TFPI-2 gene has not been investigated in glioblastoma yet. For the first time, we examined the methylation status of TFPI-2 in the tumor DNA of patients with glioblastoma. In this study, the methylation of the TFPI-2 gene promoter was detected in 22 of the 99 glioblastoma samples, but not in the normal brain specimens, which implies that the epigenetic silencing of the TFPI-2 pathway might be involved in gliomagenesis. Although the hypermethylation of the TFPI-2 promoter was frequently found in glioblastoma, it was not associated with patients’ sex and age or tumor multifocality in our series. Regarding patients’ outcome, a statistically significant association was found between the methylation of TFPI-2 and overall survival of patients. In our set of glioblastoma patients, the methylation of the TFPI-2 gene promoter was detected in 4.7% of long-term survivors with glioblastoma and in 27% of short-term survivors. The overall methylation frequency of this gene in our glioblastoma series was 22.2%, a percentage that is similar as reported in primary gastric carcinomas and lung cancers, but much lower than in nasopharyngeal carcinomas or colorectal tumors (129, 173-175). These findings suggest that the prognostic value of the promoter hypermethylation of TFPI-2 could be tissue-specific. Therefore, such findings may suggest TFPI-2 methylation as a molecular marker for glioblastoma. The restoration of TFPI-2 expression in tumor tissues inhibits invasion, tumor growth, and metastasis, which creates a novel possibility of cancer patient treatment (172). DNA methylation has shown promise as a potential biomarker for early detection, survival prognosis, or prediction of therapy response in a variety of malignancies, including glioblastoma. However, in recent years, a methylator phenotype based on concurrently methylated multiple tumor suppressor genes, also called the CIMP, is being considered to have a greater clinical value than a single gene methylation (177). Numerous studies have suggested that CIMP status might be associated with progression, recurrence, as well as long-term survival in different types of cancer, such as non–small cell lung cancer (178), acute lymphoblastic leukemia (179), neuroblastoma (180) or colon cancer (181). In glioblastoma, Noushmehr et al. detected a panel of CIMP including an 8gene signature (ANKRD43, HFE, MAL, LGALS3, FAS-1, FAS-2, RHO-F, and DOCK5) and found that patients with CIMP tumors were younger at the time of diagnosis and experienced a significantly improved outcome (8). These CIMP tumors are a subclass of the GBM proneural subtype defined by Phillips et al. and Verhaak et al. (182, 183). They were shown to be 111 associated with secondary and recurrent GBMs, IDH1 somatic mutation, younger age at diagnosis, and longer survival (5). Etcheverry et al. identified 6 CpG sites associated with overall survival; they were located in SOX10, FNDC3B, TBX3, DGKI, and FSD1 gene promoters (5). However, studies with many more cases will be needed to carefully elucidate the effects of CIMP status on survival and the most reliable panel of CIMP markers remains to be determined. Future studies are needed including prognosis as a landmark to identify these markers. In this study, we identified and characterized a distinct molecular subgroup in human glioblastomas. In our study, 6 genes (AREG, CASP8, DR4, GATA4, GATA6, and TFPI2) associated with overall survival were identified. The association between the concomitant methylation of 6 genes and patients’ survival showed a more significant effect compared with the analysis of single methylated genes. In our study, we have shown that not only methylation of an individual gene, but also comethylation of multiple genes predicts a poor prognosis in glioblastoma. Our data indicate that glioblastomas could be divided into 2 distinct subgroups with different molecular and clinical phenotypes. This molecular classification could have implications for different therapeutic strategies individually tailored to each patient with glioblastoma. This finding also raises the distinct possibility that there could be more than one type of a methylator phenotype in glioblastoma. Further studies are needed to confirm our data in a larger series of glioblastomas. 112 CONCLUSIONS 1. A decreased expression of AREG, COX7A1, KRT81, NPTX2, and SPINT1 genes in glioblastoma tissue sample in comparison with the expression of these genes in control brain samples indicates a potential role of these genes in gliomagenesis. 2. The expression of AREG and NTPX2 genes was found to be associated with their methylation status, while differences in the expression of COX7A1, KRT81, and SPINT1 genes were not related to their methylation status. 3. The investigation of 11 new epigenetic markers (AREG, CASP8, CD81, DcR1, DR4, GATA4, GATA6, hMLH1, NPTX2, and TFPI2 TES) in 100 glioblastoma samples showed tumor heterogeneity and different frequency of the promoter methylation of the analyzed genes: AREG, 59%; CASP8, 55%; CD81, 48%; DcR1, 28%; DR4, 43%; GATA4, 23%; GATA6, 68%; hMLH1, 2%; NPTX2, 53%; TES, 67%; and TFPI2, 22%. 4. CASP8 and GATA4 gene promoter methylation was found to be significantly associated with older age of patients with glioblastoma. The association between AREG and TFPI2 gene promoter methylation and patients’ age was of borderline significance. No association between the methylation status of CD81, DcR1, DR4, GATA6, hMLH1, NPTX2, and TES genes and patients’ age was found. The methylation status of none of these analyzed genes was significantly associated with gender. The methylation status of only one gene – NPTX2 – was significantly associated with tumor multifocality. 5. The methylation status of the following 4 gene promoters was significantly related to patients’ survival after surgery: AREG, CASP8, GATA6, and TFPI2. Identification of the methylation status of these genes could be one of the objective criteria in the prognosis of disease course in patients with glioblastoma and could supplement the list of already known epigenetic markers. 6. The methylation status of a combination of 6 genes (AREG, CASP8, DR4, GATA4, GATA6, and TFPI2) was found to be a more accurate independent prognostic factor for patients’ survival after surgery as compared with the methylation status of individual genes. The molecular classification of glioblastomas according to the methylation profile of a combination of these 6 genes could help clinicians tailor an appropriate treatment strategy. 113 REFERENCES 1. Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, et al. Long-term survival with glioblastoma multiforme. Brain 2007;130:2596-606. 2. Martinez R, Martin-Subero JI, Rohde V, Kirsch M, Alaminos M, Fernandez AF, et al. A microarray-based DNA methylation study of glioblastoma multiforme. Epigenetics 2009;4:255-64. 3. Rahnenführer J, Beerenwinkel N, Schulz WA, Hartmann C, von Deimling A, Wullich B, et al. Estimating cancer survival and clinical outcome based on genetic tumor progression scores. Bioinformatics 2008;21:2438-46. 4. Poplawski T, Tomaszewska K, Galicki M, Morawiec Z, Blasiak J. Promoter methylation of cancer-related genes in gastric carcinoma. Exp Oncol 2008;30(2):112-6. 5. Etcheverry A, Aubry M, de Tayrac M, Vauleon E, Boniface R, Guenot F, et al. DNA methylation in glioblastoma: impact on gene expression and clinical outcome. BMC Genomics 2010;11:701. 6. Laffaire J, Everhard S, Idbaih A, Crinière E, Marie Y, de Reyniès A, et al. Methylation profiling identifies 2 groups of gliomas according to their tumorigenesis. Neuro-Oncology 2011;13:84-98. 7. Mueller W, Nutt CL, Ehrich M, Riemenschneider MJ, von Deimling A, van den Boom D, et al. Downregulation of RUNX3 and TES by hypermethylation in glioblastoma. Oncogene 2007;26:583-93. 8. Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP, et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 2010;17:510-22. 9. Di Vinci A, Casciano I, Marasco E, Banelli B, Ravetti GL, Borzì L, et al. Quantitative methylation analysis of HOXA3, 7, 9, and 10 genes in glioma: association with tumor WHO grade and clinical outcome. J Cancer Res Clin Oncol 2012;138(1):35-47. 10. Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC, et al. The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol 2002;61(3):215-25. 11. Agarwal S, Sane R, Oberoi R, Ohlfest JR, Elmquist WF. Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain. Expert Rev Mol Med 2011;13:e17. 12. Weller M, Stupp R, Hegi M, Wick W. Individualized targeted therapy for glioblastoma: fact or fiction? Cancer J 2012;18(1):40-4. 114 13. Lim SK, Llaguno SR, McKay RM, Parada LF. Glioblastoma multiforme: a perspective on recent findings in human cancer and mouse models. BMB Rep 2011;44(3):158-64. 14. Preusser M, Haberler C, Hainfellner JA. Malignant glioma: neuropathology and neurobiology. Wien Med Wochenschr 2006;156(1112):332-7. 15. Rock K, McArdle O, Forde P, Dunne M, Fitzpatrick D, O'Neill B, et al. A clinical review of treatment outcomes in glioblastoma multiforme--thevalidation in a non-trial population of the results of a randomised PhaseIII clinical trial: has a more radical approach improved survival? Br J Radiol 2012;85 (1017):729-33. 16. Kanu OO, Mehta A, Di C, Lin N, Bortoff K, Bigner DD, et al. Glioblastoma multiforme: a review of therapeutic targets. Expert Opin Ther Targets 2009;13(6):701-18. 17. Knights MJ, Kyle S, Ismail A. Characteristic features of stem cells in glioblastomas: from cellular biology to genetics. Brain Pathol 2012;22 (5):592-6006. 18. Hong B, Wiese B, Bremer M, Heissler HE, Heidenreich F, Krauss JK, et al. Multiple microsurgical resections for repeated recurrence of glioblastoma multiforme. Am J Clin Oncol 2012 [Epub ahead of print]. 19. Martinez R, Esteller M. The DNA methylome of glioblastoma multiforme. Neurobiol Dis 2010;39(1):40-6. 20. Kang SH, Yu MO, Park KJ, Chi SG, Park DH, Chung YG. Activated STAT3 regulates hypoxia-induced angiogenesis and cell migration in human glioblastoma. Neurosurgery 2010;67(5):1386-95. 21. Huang Z, Cheng L, Guryanova OA, Wu Q, Bao S. Cancer stem cells in glioblastoma--molecular signaling and therapeutic targeting. Protein Cell 2010;1(7):638-55. 22. Stopschinski BE, Beier CP, Beier D. Glioblastoma cancer stem cells - From concept to clinical application. Cancer Lett 2012 [Epub ahead of print]. 23. Wei Zhang, Gregory N. Fuller, editor. Genomic And Molecular Neuro-Oncology: Jones & Bartlett Learning; 2003. 24. Brennan C. Genomic profiles of glioma. Curr Neurol Neurosci Rep 2011;11(3):291-7. 25. Gupta R, Webb-Myers R, Flanagan S, Buckland ME. Isocitrate dehydrogenase mutations in diffuse gliomas: clinical and aetiological implications. J Clin Pathol 2011;64(10):835-44. 26. Martinez R, Schackert G, Yaya-Tur R, Rojas-Marcos I, Herman JG, Esteller M. Frequent hypermethylation of the DNA repair gene 115 MGMT in long-term survivors of glioblastoma multiforme. J Neurooncol 2007;83(1):91-3. 27. Das P, Puri T, Jha P, Pathak P, Joshi N, Suri V, et al. A clinicopathological and molecular analysis of glioblastoma multiforme with long-term survival. J Clin Neurosci 2011;18(1):66-70. 28. Lamborn KR, Chang SM, Prados MD. Prognostic factors for survival of patients with glioblastoma: recursive partitioning analysis. Neuro Oncol 2004;6(3):227-35. 29. Barbus S, Tews B, Karra D, Hahn M, Radlwimmer B, Delhomme N, et al. Differential retinoic acid signaling in tumors of longand short-term glioblastoma survivors. J Natl Cancer Inst 2011;103(7):598606. 30. Tost J, editor. DNA methylation: methods and protocols: Humana Press; 2008. 31. Tollefsbol T, editor. Handbook of epigenetics: the new molecular and medical genetics: Elsevier; 2011. 32. Maxwell A, McCudden CR, Wians F, Willis MS. Recent advances in the detection of prostate cancer using epigenetic markers in commonly collected laboratory samples. Labmedicine 2009;40(3):171-78. 33. Nagarajan RP, Costello JF. Epigenetic mechanisms in glioblastoma multiforme. Semin Cancer Biol 2009;19(3):188-97. 34. Gal-Yam EN, Saito Y, Egger G, Jones PA. Cancer epigenetics: modifications, screening, and therapy. Annu Rev Med 2008;59:267-80. 35. Das PM, Singal R. DNA methylation and cancer. J Clin Oncol 2004;22(22):4632-42. 36. von Deimling A, Korshunov A, Hartmann C. The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and IDH1 mutations. Brain Pathol 2011;21(1):74-87. 37. Gonzalez-Gomez P, Bello MJ, Arjona D, Lomas J, Alonso ME, De Campos JM, et al. Promoter hypermethylation of multiple genes in astrocytic gliomas. Int J Oncol 2003;22(3):601-8. 38. Uhlmann K, Rohde K, Zeller C, Szymas J, Vogel S, Marczinek K, et al. Distinct methylation profiles of glioma subtypes. Int J Cancer 2003;106(1):52-9. 39. Yu J, Zhang H, Gu J, Lin S, Li J, Lu W, et al. Methylation profiles of thirty four promoter-CpG islands and concordant methylation behaviours of sixteen genes that may contribute to carcinogenesis of astrocytoma. BMC Cancer 2004;4:65. 40. Hamilton JP. Epigenetics: principles and practice. Dig Dis 2011;29(2):130-5. 116 41. Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nat Biotechnol 2010;28(10):1069-78. 42. Tsai HC, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res 2011;21(3):502-17. 43. Meador JA, Su Y, Ravanat JL, Balajee AS. DNA-dependent protein kinase (DNA-PK)-deficient human glioblastoma cells are preferentially sensitized by Zebularine. Carcinogenesis 2010;31(2):184-91. 44. Williams K, Christensen J, Helin K. DNA methylation. TET proteins-guardians of CpG islands? EMBO Rep 2012;13:28–35. 45. Münzel M, Globisch D, Carell T. 5-Hydroxymethylcytosine, the sixth base of the genome. Angew Chem Int Ed Engl 2011;50(29):64608. 46. Branco MR, Ficz G, Reik W. Uncovering the role of 5hydroxymethylcytosine in the epigenome. Nat Rev Genet 2012;13:7-13. 47. Kreth S, Thon N, Kreth FW. Epigenetics in human gliomas. Cancer Lett 2012 [Epub ahead of print]. 48. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352(10):997-1003. 49. Elias A, Siegelin MD, Steinmüller A, von Deimling A, Lass U, Korn B, et al. Epigenetic silencing of death receptor 4 mediates tumor necrosis factor-related apoptosis-inducing ligand resistance in gliomas. Clin Cancer Res 2009;15:5457-65. 50. Hervouet E, Vallette FM, Cartron PF. Impact of the DNA methyltransferases expression on the methylation status of apoptosisassociated genes in glioblastoma multiforme. Cell Death Dis 2010; 1:e8. 51. Cecener G, Tunca B, Egeli U, Bekar A, Tezcan G, Erturk E, et al. The promoter hypermethylation status of GATA6, MGMT, and FHIT in glioblastoma. Cell Mol Neurobiol 2012;32(3):237-44. 52. Kamnasaran D, Qian B, Hawkins C, Stanford WL, Guha A. GATA6 is an astrocytoma tumor suppressor gene identified by gene trapping of mouse glioma model. Proc Natl Acad Sci USA 2007;104(19):8053-8. 53. Felsberg J, Thon N, Eigenbrod S, Hentschel B, Sabel MC, Westphal M, et al. Promoter methylation and expression of MGMT and the DNA mismatch repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and recurrent glioblastomas. Int J Cancer 2011;129:659-70. 54. Fukushima T, Katayama Y, Watanabe T, Yoshino A, Ogino A, Ohta T, et al. Promoter hypermethylation of mismatch repair gene HMLH1 predicts the clinical response of malignant astrocytomas to nitrosourea. Clin Cancer Res 2005;11:1539-44. 117 55. Plowman GD, Green JM, McDonald VL, Neubauer MG, Disteche CM, Todaro GJ, et al. The amphiregulin gene encodes a novel epidermal growth factor-related protein with tumor-inhibitory activity. Mol Cell Biol 1990;10(5):1969-81. 56. Mallakin A, Sugiyama T, Kai F, Taneja P, Kendig RD, Frazier DP, et al. The Arf-inducing transcription factor Dmp1 encodes a transcriptional activator of amphiregulin, thrombospondin-1, JunB and Egr1. Int J Cancer 2010;126(5):1403-16. 57. Busser B, Sancey L, Brambilla E, Coll JL, Hurbin A. The multiple roles of amphiregulin in human cancer. Biochim Biophys Acta 2011;1816(2):119-31. 58. Yamashita S, Tsujino Y, Moriguchi K, Tatematsu M, Ushijima T. Chemical genomic screening for methylation-silenced genes in gastric cancer cell lines using 5-aza-2'-deoxycytidine treatment and oligonucleotide microarray. Cancer Sci 2006;97(1):64-71. 59. Shih IeM, Chen L, Wang CC, Gu J, Davidson B, Cope L, et al. Distinct DNA methylation profiles in ovarian serous neoplasms and their implications in ovarian carcinogenesis. Am J Obstet Gynecol 2010;203(6):584. 60. Liang G, Gonzales FA, Jones PA, Orntoft TF, Thykjaer T. Analysis of gene induction in human fibroblasts and bladder cancer cells exposed to the methylation inhibitor 5-aza-2'-deoxycytidine. Cancer Res 2002;62(4):961-6. 61. Database tGhg. Available from: URL: http://www.genecards.org/cgi-bin/carddisp.pl?gene=CASP8 62. Qi L, Bellail AC, Rossi MR, Zhang Z, Pang H, Hunter S, et al. Heterogeneity of primary glioblastoma cells in the expression of caspase-8 and the response to TRAIL-induced apoptosis. Apoptosis 2011;16(11):1150-64. 63. Fulda S, Debatin KM. 5-Aza-2'-deoxycytidine and IFNgamma cooperate to sensitize for TRAIL-induced apoptosis by upregulating caspase-8. Oncogene 2006;25(37):5125-33. 64. Khaider NG, Lane D, Matte I, Rancourt C, Piché A. Targeted ovarian cancer treatment: the TRAILs of resistance. Am J Cancer Res 2012;2(1):75-92. 65. NCBI. The National Center for Biotechnology Information Available from: URL: http://www.ncbi.nlm.nih.gov/gene?db=gene&cmd=Retrieve&dopt=full_rep ort&list_uids=975 118 66. Mazzocca A, Liotta F, Carloni V. Tetraspanin CD81-regulated cell motility plays a critical role in intrahepatic metastasis of hepatocellular carcinoma. Gastroenterology 2008;135(1):244-56. 67. Schöniger-Hekele M, Hänel S, Wrba F, Müller C. Hepatocellular carcinoma – survival and clinical characteristics in relation to various histologic molecular markers in Western patients. Liver Int 2005;25(1):62-9. 68. Drucker L, Tohami T, Tartakover-Matalon S, Zismanov V, Shapiro H, Radnay J, et al. Promoter hypermethylation of tetraspanin members contributes to their silencing in myeloma cell lines. Carcinogenesis 2006;27(2):197-204. 69. Hemler ME. Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol 2005;6(10):801-11. 70. COX7A1 cytochrome c oxidase subunit VIIa polypeptide 1 (muscle) [Homo sapiens]. Available from: URL: http://www.ncbi.nlm.nih.gov/gene/1346 71. Rönn T, Poulsen P, Hansson O, Holmkvist J, Almgren P, Nilsson P, et al. Age influences DNA methylation and gene expression of COX7A1 in human skeletal muscle. Diabetologia 2008;51(7):1159-68. 72. Hill VK, Ricketts C, Bieche I, Vacher S, Gentle D, Lewis C, et al. Genome-wide DNA methylation profiling of CpG islands in breast cancer identifies novel genes associated with tumorigenicity. Cancer Res 2011;71(8):2988-99. 73. Azhikina T, Gainetdinov I, Skvortsova Y, Sverdlov E. Methylation-free site patterns along a 1-Mb locus on Chr19 in cancerous and normal cells are similar. A new fast approach for analyzing unmethylated CCGG sites distribution. Mol Genet Genomics 2006; 275(6):615-22. 74. Tabarés-Seisdedos R, Rubenstein JL. Chromosome 8p as a potential hub for developmental neuropsychiatric disorders: implications for schizophrenia, autism and cancer. Mol Psychiatry 2009;14(6):563-89. 75. Wemmert S, Ketter R, Rahnenführer J, Beerenwinkel N, Strowitzki M, Feiden W, et al. Patients with high-grade gliomas harboring deletions of chromosomes 9p and 10q benefit from temozolomide treatment. Neoplasia 2005;7(10):883-93. 76. Lee KH, Lim SW, Kim HG, Kim DY, Ryu SY, Joo JK, et al. Lack of death receptor 4 (DR4) expression through gene promoter methylation in gastric carcinoma. Langenbecks Arch Surg 2009;394(4):66170. 119 77. TNFRSF10C tumor necrosis factor receptor superfamily, member 10c, decoy without an intracellular domain [Homo sapiens] Available from: URL: http://www.ncbi.nlm.nih.gov/gene/8794 78. Tserga A, Michalopoulos NV, Levidou G, Korkolopoulou P, Zografos G, Patsouris E, et al. Association of aberrant DNA methylation with clinicopathological features in breast cancer. Oncol Rep 2012;27(5):1630-8. 79. Dauksa A, Gulbinas A, Barauskas G, Pundzius J, Oldenburg J, El-Maarri O. Whole blood DNA aberrant methylation in pancreatic adenocarcinoma shows association with the course of the disease: a pilot study. PLoS One 2012; 7(5):e37509. 80. Shin SH, Kim BH, Jang JJ, Suh KS, Kang GH. Identification of novel methylation markers in hepatocellular carcinoma using a methylation array. J Korean Med Sci 2010;25(8):1152-9. 81. TNFRSF10A tumor necrosis factor receptor superfamily, member 10a [Homo sapiens]. Available from: URL: http://www.ncbi.nlm.nih.gov/gene/8797 82. van Noesel MM, van Bezouw S, Voûte PA, Herman JG, Pieters R, Versteeg R. Clustering of hypermethylated genes in neuroblastoma. Genes Chromosomes Cancer 2003;38(3):226-33. 83. Shivapurkar N, Toyooka S, Toyooka KO, Reddy J, Miyajima K, Suzuki M, et al. Aberrant methylation of trail decoy receptor genes is frequent in multiple tumor types. Int J Cancer 2004;109(5):786-92. 84. GATA4 GATA binding protein 4 [Homo sapiens]. Available from: URL: http://www.ncbi.nlm.nih.gov/gene/2626 85. Suzuki YJ. Cell signaling pathways for the regulation of GATA4 transcription factor: implications for cell growth and apoptosis. Cell Signal 2011;23(7):1094-9. 86. Agnihotri S, Wolf A, Picard D, Hawkins C, Guha A. GATA4 is a regulator of astrocyte cell proliferation and apoptosis in the human and murine central nervous system. Oncogene 2009;28(34):3033-46. 87. Kuo CT, Morrisey EE, Anandappa R, Sigrist K, Lu MM, Parmacek MS, et al. GATA4 transcription factor is required for ventral morphogenesis and heart tube formation. Genes Dev 1997;11(8):1048-60. 88. Reamon-Buettner SM, Cho SH, Borlak J. Mutations in the 3'untranslated region of GATA4 as molecular hotspots for congenital heart disease (CHD). BMC Med Genet 2007;8(38). 89. Akiyama Y, Watkins N, Suzuki H, Jair KW, van Engeland M, Esteller M, et al. GATA-4 and GATA-5 transcription factor genes and potential downstream antitumor target genes are epigenetically silenced in colorectal and gastric cancer. Mol Cell Biol 2003;23(23):8429-39. 120 90. Guo M, Akiyama Y, House MG, Hooker CM, Heath E, Gabrielson E, et al. Hypermethylation of the GATA genes in lung cancer. Clin Cancer Res 2004;10(23):7917-24. 91. Guo M, House MG, Akiyama Y, Qi Y, Capagna D, Harmon J, et al. Hypermethylation of the GATA gene family in esophageal cancer. Int J Cancer 2006;19(9):2078-83. 92. Caslini C, Capo-chichi CD, Roland IH, Nicolas E, Yeung AT, Xu XX. Histone modifications silence the GATA transcription factor genes in ovarian cancer. Oncogene 2006;25(39):5446-61. 93. Hua G, Zhu B, Rosa F, Deblon N, Adélaïde J, Kahn-Perlès B, et al. A negative feedback regulatory loop associates the tyrosine kinase receptor ERBB2 and the transcription factor GATA4 in breast cancer cells. Mol Cancer Res 2009;7(3):402-14. 94. Agnihotri S, Wolf A, Munoz DM, Smith CJ, Gajadhar A, Restrepo A, et al. A GATA4-regulated tumor suppressor network represses formation of malignant human astrocytomas. J Exp Med 2011;208(4):689702. 95. Zheng R, Blobel GA. GATA transcription factors and cancer. Genes Cancer 2010;1(12):1178-88. 96. Lowry JA, Atchley WR. Molecular evolution of the GATA family of transcription factors: conservation within the DNA-binding domain. J Mol Evol 2000;50(2):103-15. 97. Wheeler JM, Bodmer WF, Mortensen NJ. DNA mismatch repair genes and colorectal cancer. Gut 2000;47(1):148-53. 98. GeneCards. hMLH1. Available from: URL: http://www.genecards.org/cgi-bin/carddisp.pl?gene=MLH1&search=hMLH1 99. Deguchi M, Shiina H, Igawa M, Kaneuchi M, Nakajima K, Dahiya R. DNA mismatch repair genes in renal cell carcinoma. J Urol 2003;169(6):2365-71. 100. Deng G, Peng E, Gum J, Terdiman J, Sleisenger M, Kim YS. Methylation of hMLH1 promoter correlates with the gene silencing with a region-specific manner in colorectal cancer. Br J Cancer 2002;86(4):574-9. 101. Wani M, Afroze D, Makhdoomi M, Hamid I, Wani B, Bhat G, et al. Promoter Methylation Status of DNA Repair Gene (hMLH1) in Gastric Carcinoma Patients of the Kashmir Valley. Asian Pac J Cancer Prev 2012;13(8):4177-81. 102. Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, et al. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci USA 1998;95(12):6870-5. 121 103. Xinarianos G, Liloglou T, Prime W, Maloney P, Callaghan J, Fielding P, et al. hMLH1 and hMSH2 expression correlates with allelic imbalance on chromosome 3p in non-small cell lung carcinomas. Cancer Res 2000;60(15):4216-21. 104. KRT81 keratin 81 [Homo sapiens]. Available from: URL: http://www.ncbi.nlm.nih.gov/gene/3887 105. Moll R, Franke WW, Schiller DL, Geiger B, Krepler R. The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells. Cell 1982;31(1):11-24. 106. Moll R, Divo M, Langbein L. The human keratins: biology and pathology. Histochem Cell Biol 2008;129(6):705-33. 107. Campayo M, Navarro A, Viñolas N, Tejero R, Muñoz C, Diaz T, et al. A dual role for KRT81: a miR-SNP associated with recurrence in non-small-cell lung cancer and a novel marker of squamous cell lung carcinoma. PLoS One. 2011;6(7) e22509. 108. NPTX2 neuronal pentraxin II [Homo sapiens]. Available from: URL: http://www.ncbi.nlm.nih.gov/gene/4885 109. Bocklandt S, Lin W, Sehl ME, Sánchez FJ, Sinsheimer JS, Horvath S, et al. Epigenetic predictor of age. PLoS One 2011;6(6) e14821. 110. Koch CM, Wagner W. Epigenetic-aging-signature to determine age in different tissues. Aging (Albany NY) 2011;3(10):1018-27. 111. Sato N, Fukushima N, Maitra A, Matsubayashi H, Yeo CJ, Cameron JL, et al. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res 2003;63(13):3735-42. 112. Zhang L, Gao J, Li L, Li Z, Du Y, Gong Y. The neuronal pentraxin II gene (NPTX2) inhibit proliferation and invasion of pancreatic cancer cells in vitro. Mol Biol Rep 2011;38(8):4903-11. 113. Zhang Y, Yang B, Du Z, Gao YT, Wang YJ, Jing X, et al. Identification and validation of specific methylation profile in bile for differential diagnosis of malignant biliary stricture. Clin Biochem 2010;43(16):1340-4. 114. Enjuanes A, Fernàndez V, Hernández L, Navarro A, Beà S, Pinyol M, et al. Identification of methylated genes associated with aggressive clinicopathological features in mantle cell lymphoma. PLoS One 2011;6(5) e19736. 115. Haematology AoGaCiOa. SPINT1. Available from: URL: http://atlasgeneticsoncology.org/Genes/SPINT1ID44384ch15q15.html 116. TES testis derived transcript (3 LIM domains) [Homo sapiens]. Available from: URL: http://www.ncbi.nlm.nih.gov/gene/26136; 2011. 122 117. Tatarelli C, Linnenbach A, Mimori K, Croce CM. Characterization of the human TESTIN gene localized in the FRA7G region at 7q31.2. Genomics 2000;68(1):1-12. 118. Tobias ES, Hurlstone AF, MacKenzie E, McFarlane R, Black DM. The TES gene at 7q31.1 is methylated in tumours and encodes a novel growth-suppressing LIM domain protein. Oncogene 2001;20(22):2844-53. 119. Drusco A, Zanesi N, Roldo C, Trapasso F, Farber JL, Fong LY, et al. Knockout mice reveal a tumor suppressor function for Testin. Proc Natl Acad Sci USA 2005;102(31):10947-51. 120. Sarti M, Sevignani C, Calin GA, Aqeilan R, Shimizu M, Pentimalli F, et al. Adenoviral transduction of TESTIN gene into breast and uterine cancer cell lines promotes apoptosis and tumor reduction in vivo. Clin Cancer Res 2005;11(2):806-13. 121. Weeks RJ, Kees UR, Song S, Morison IM. Silencing of TESTIN by dense biallelic promoter methylation is the most common molecular event in childhood acute lymphoblastic leukaemia. Mol Cancer 2010;9:163. 122. GeneCards. TFPI2. Available from URL: http://www.genecards.org/cgi-bin/carddisp.pl?gene=TFPI2&search=TFPI-2 123. Werb Z. ECM and cell surface proteolysis: regulating cellular ecology. Cell 1997;91(4):439-42. 124. Valente P, Fassina G, Melchiori A, Masiello L, Cilli M, Vacca A, et al. TIMP-2 over-expression reduces invasion and angiogenesis and protects B16F10 melanoma cells from apoptosis. Int J Cancer 1998;75(2):246-53. 125. Miyagi Y, Koshikawa N, Yasumitsu H, Miyagi E, Hirahara F, Aoki I, et al. cDNA cloning and mRNA expression of a serine proteinase inhibitor secreted by cancer cells: identification as placental protein 5 and tissue factor pathway inhibitor-2. J Biochem 1994;116(5):939-42. 126. Wojtukiewicz MZ, Sierko E, Zimnoch L, Kozlowski L, Kisiel W. Immunohistochemical localization of tissue factor pathway inhibitor-2 in human tumor tissue. Thromb Haemost 2003;90(1):140-6. 127. Wong CM, Ng YL, Lee JM, Wong CC, Cheung OF, Chan CY, et al. Tissue factor pathway inhibitor-2 as a frequently silenced tumor suppressor gene in hepatocellular carcinoma. Hepatology 2007;45(5):112938. 128. Ran Y, Pan J, Hu H, Zhou Z, Sun L, Peng L, et al. A novel role for tissue factor pathway inhibitor-2 in the therapy of human esophageal carcinoma. Hum Gene Ther 2009;20(1):41-9. 123 129. Wang S, Xiao X, Zhou X, Huang T, Du C, Yu N, et al. TFPI-2 is a putative tumor suppressor gene frequently inactivated by promoter hypermethylation in nasopharyngeal carcinoma. BMC Cancer 2010;10:671. 130. Gessler F, Voss V, Seifert V, Gerlach R, Kögel D. Knockdown of TFPI-2 promotes migration and invasion of glioma cells. Neurosci Lett 2011;497(1):49-54. 131. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 2007;114(2):97-109. 132. Frommer M, McDonald LE, Millar DS, Collis CM, Watt F, Grigg GW, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci USA 1992;89(5):1827-31. 133. Herman JG, Graff JR, Myöhänen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996;93(18):9821-6. 134. Yamashita K, Upadhyay S, Osada M, Hoque MO, Xiao Y. Pharmacologic unmasking of epigenetically silenced tumor suppressor genes in esophageal squamous cell carcinoma. Cancer Cell 2002;2:485-95. 135. Lodygin D, Epanchintsev A, Menssen A, Diebold J, Hermeking H. Functional epigenomics identifies genes frequently silenced in prostate cancer. Cancer Res 2005;65:4218-27. 136. Raynal NJ, Si J, Taby RF, Gharibyan V, Ahmed S. DNA methylation does not stably lock gene expression but instead serves as a molecular mark for gene silencing memory. Cancer Res 2012;72:1170-81. 137. Pruitt K, Zinn RL, Ohm JE, McGarvey KM, Kang SH. Inhibition of SIRT1 reactivates silenced cancer genes without loss of promoter DNA hypermethylation. PLoS Genet 2006;2:e40. 138. Harrington MA, Jones PA, Imagawa M, Karin M. Cytosine methylation does not affect binding of transcription factor Sp1. Proc Natl Acad Sci USA 1988;85:2066-70. 139. Smith KT, Coffee B, Reines D. Occupancy and synergistic activation of the FMR1 promoter by Nrf-1 and Sp1 in vivo. Hum Mol Genet 2004;13:1611-21. 140. Lamber EP, Horwitz AA, Parvin JD. R BRCA1 represses amphiregulin gene expression. Cancer Research 2010;70:996-1005. 141. Shoyab M, McDonald VL, Bradley JG, Todaro GJ. Amphiregulin: a bifunctional growth-modulating glycoprotein produced by the phorbol 12-myristate 13-acetate-treated human breast adenocarcinoma cell line MCF-7. Proc Natl Acad Sci USA 1988;85(17):6528-32. 124 142. Duhem-Tonnelle V, Bièche I, Vacher S, Loyens A, Maurage CA, Collier F, et al. Differential distribution of ErbB receptors in human glioblastoma multiforme: expression of ErbB3 in CD133-positive putative cancer stem cells. J Neuropathol Exp Neurol. 2010;69:606-22. 143. Willmarth NE, Ethier SP. Amphiregulin as a novel target for breast cancer therapy. J Mammary Gland Biol Neoplasia 2008;13:171-9. 144. Ebert M, Yokoyama M, Kobrin MS, Friess H, Lopez ME, Buchler MW, et al. Induction and expression of amphiregulin in human pancreatic cancer. Cancer Research 1994;54:3959-62. 145. Fontanini G, De Laurentiis M, Vignati S, Chinè S, Lucchi M, Silvestri V, et al. Evaluation of epidermal growth factor-related growth factors and receptors and of neoangiogenesis in completely resected stage IIIIA non-small-cell lung cancer: amphiregulin and microvessel count are independent prognostic indicators of survival. Clin Cancer Res 1998;4:2419. 146. Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol 1995;19:183-232. 147. Cunningham JM, Christensen ER, Tester DJ, Kim CY, Roche PC, Burgart LJ, et al. Hypermethylation of the HMLH1 promoter in colon cancer with microsatellite instability. Cancer Research 1998;58:345-3460. 148. Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Human Molecular Genetics 2007;16:50-9. 149. Kane MF, Loda M, Gaida GM, Lipman J, Mishra R, Goldman H, et al. Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repairdefective human tumor cell lines. Cancer Research 1997;57:808-11. 150. Leung SY, Yuen ST, Chung LP, Chu KM, Chan AS, Ho JC. hMLH1 promoter methylation and lack of hMLH1 expression in sporadic gastric carcinomas with high-frequency microsatellite instability. Cancer Research 1999;59:159-64. 151. Kanamori M, Kon H, Nobukuni T, Nomura S, Sugano K, Mashiyama S, et al. Microsatellite instability and the PTEN1 gene mutation in a subset of early onset gliomas carrying germline mutation or promoter methylation of the hMLH1 gene. Oncogene 2000;19:1564-71. 152. Friedman HS, McLendon RE, Kerby T, Dugan M, Bigner SH, Henry AJ, et al. DNA mismatch repair and O6-alkylguanine-DNA alkyltransferase analysis and response to temodal in newly diagnosed malignant glioma. J Clin Oncol 1998;16:3851-7. 125 153. Hsu YC, Perin MS. Human neuronal pentraxin II (NPTX2): conservation, genomic structure, and chromosomal localization. Genomics 1995;28:220-7. 154. Carlson MR, Pope WB, Horvath S, Braunstein JG, Nghiemphu P, Tso CL, et al. Relationship between survival and edema in malignant gliomas: role of vascular endothelial growth factor and neuronal pentraxin 2. Clinical Cancer Research 2007;13:2592-8. 155. Gunduz EG, Gunduz M, Beder L, Nagatsuka H, Fukushima K, Sutcu R, et al. Downregulation of TESTIN and its association with cancer history and a tendency toward poor survival in head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 2009;135:254-60. 156. Ma H, Weng D, Chen Y, Huang W, Pan K, Wang H, et al. Extensive analysis of D7S486 in primary gastric cancer supports TESTIN as a candidate tumor suppressor gene. Molecular Cancer 2010;9:190. 157. Shureiqi I, Jiang W, Fischer SM, Xu X, Chen D, Lee JJ, et al. GATA-6 transcriptional regulation of 15-lipoxygenase-1 during NSAIDinduced apoptosis in colorectal cancer cells. Cancer Res 2002;62:1178-83. 158. Haveri H, Westerholm-Ormio M, Lindfors K, Maki M, Savilahti E, Andersson L, et al. Transcription factors GATA-4 and GATA-6 in normal and neoplastic human gastrointestinal mucosa. BMC Gastroenterol 2008;8:9. 159. Levy S, Todd SC, Maecker HT. CD81 (TAPA-1): a molecule involved in signal transduction and cell adhesion in the immune system. Annu Rew Immunol 1998; 16:89-109. 160. Sadikovic B, Yoshimoto M, Chilton-Macneill, Thorner P, Squire JA, Zielenska M. Identification of interactive networks of gene expression associated with osteosarcoma oncogenesis by integrated molecular profiling. Hum Mol Genet 2009; 18(11):1962-75. 161. Bae SI, Cheriyath V, Jacobs BS, Reu FJ, Borden EC. Reversal of methylation silencing of Apo2L/TRAIL receptor 1 (DR4) expression overcomes resistance of SK-MEL-3 and SK-MEL-28 melanoma cells to interferons (IFNs) or Apo2L/TRAIL. Oncogene 2008;27:490-8. 162. Aguilera DG, Das CM, Sinnappah-Kang ND, Joyce C, Taylor PH, Wen S, et al. Reactivation of death receptor 4 (DR4) expression sensitizes medulloblastoma cell lines to TRAIL. J Neurooncol 2009;93:30318. 163. Hellebrekers DM, Lentjes MH, van den Bosch SM, Melotte V, Wouters KA, Daenen KL, et al. GATA4 and GATA5 are potential tumor suppressors and biomarkers in colorectal cancer. Clin Cancer Res 2009; 15(12):3990-7. 126 164. Wen XZ, Akiyama Y, Pan KF, Liu ZJ, Lu ZM, Zhou J, et al. Methylation of GATA-4 and GATA-5 and development of sporadic gastric carcinomas. World J Gastroenterol 2010;16(10):1201-8. 165. Teodoridis JM, Hall J, Marsh S, Kannall HD, Smyth C, Curto J, et al. CpG island methylation of DNA damage response genes in advanced ovarian cancer. Cancer Res 2005;65(19):8961-7. 166. Cheng Y, Kim JW, Liu W, Dunn TA, Luo J, Loza MJ, et al. Genetic and epigenetic inactivation of TNFRSF10C in human prostate cancer. Prostate 2009;69(3):327-35. 167. Coniglio SJ, Eugenin E, Dobrenis K, Stanley ER, West BL, Symons MH, et al. Microglial stimulation of glioblastoma invasion involves epidermal growth factor receptor (EGFR) and colony stimulating factor 1 receptor (CSF-1R) signaling. Mol Med 2012;18:519-27. 168. Westermarck J, Kähäri VM. Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 1999;13(8):78192. 169. Park JB, Kwak HJ, Lee SH. Role of hyaluronan in glioma invasion. Cell Adh Migr 2008;2(3):202-7. 170. Ran Y, Pan J, Hu H, Zhou Z, Sun L, Peng L, et al. A novel role for tissue factor pathway inhibitor-2 in the therapy of human esophageal carcinoma. Hum Gene Ther 2009;20(1):41-9. 171. Konduri SD, Srivenugopal KS, Yanamandra N, Dinh DH, Olivero WC, Gujrati M, et al. Promoter methylation and silencing of the tissue factor pathway inhibitor-2 (TFPI-2), a gene encoding an inhibitor of matrix metalloproteinases in human glioma cells. Oncogene 2003;22(29):4509-16. 172. Sierko E, Wojtukiewicz MZ, Kisiel W. The role of tissue factor pathway inhibitor-2 in cancer biology. Semin Thromb Hemost 2007;33(7):653-9. 173. Hibi K, Goto T, Shirahata A, Saito M, Kigawa G, Nemoto H, et al. Detection of TFPI2 methylation in the serum of colorectal cancer patients. Cancer Lett 2011;311(1):96-100. 174. Wu D, Xiong L, Wu S, Jiang M, Lian G, Wang M. TFPI-2 methylation predicts poor prognosis in non-small cell lung cancer. Lung Cancer 2012;76(1):106-11. 175. Hibi K, Goto T, Kitamura YH, Sakuraba K, Shirahata A, Mizukami H. Methylation of the TFPI2 gene is frequently detected in advanced gastric carcinoma. Anticancer Res 2010;30(10):4131-3. 176. Sato N, Parker AR, Fukushima N, Miyagi Y, IacobuzioDonahue CA, Eshleman JR, et al. Epigenetic inactivation of TFPI-2 as a 127 common mechanism associated with growth and invasion of pancreatic ductal adenocarcinoma. Oncogene 2005;24(4):850-8. 177. Tanemura A, Terando AM, Sim MS, van Hoesel AQ, de Maat MF, Morton DL, et al. CpG island methylator phenotype predicts progression of malignant melanoma. Clin Cancer Res 2009;15(5):1801-7. 178. Liu Z, Zhao J, Chen XF, Li W, Liu R, Lei Z, et al. CpG island methylator phenotype involving tumor suppressor genes located on chromosome 3p in non-small cell lung cancer. Lung Cancer 2008;62(1):1522. 179. Roman-Gomez J, Jimenez-Velasco A, Agirre X, Prosper F, Heiniger A, Torres A. Lack of CpG island methylator phenotype defines a clinical subtype of T-cell acute lymphoblastic leukemia associated with good prognosis. J Clin Oncol 2005;23(28):7043-9. 180. Abe M, Ohira M, Kaneda A, Yagi Y, Yamamoto S, Kitano Y, et al. CpG island methylator phenotype is a strong determinant of poor prognosis in neuroblastomas. Cancer Res 2005;65(3):828-34. 181. Ogino S, Nosho K, Kirkner GJ, Kawasaki T, Meyerhardt JA, Loda M, et al. CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut 2009;58(1):90-6. 182. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 2006;9(3):157-73. 183. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al; Cancer Genome Atlas Research Network. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17(1):98-110. 128 LIST OF PUBLICATIONS 1. Vaitkienė, Paulina; Skiriutė, Daina; Skauminas, Kęstutis; Tamašauskas, Arimantas. GATA4 and DcR1 methylation in glioblastomas // Diagnostic pathology [electronic resource]. [London] : BioMed Central. ISSN 1746-1596. 2013, vol. 8, no. 1, 1 <http://www.ncbi.nlm.nih.gov/pubmed/23320456>. [ISI Master journal list; MEDLINE; CAS; Index Copernicus; Scopus]. [Citav. rod.: 1,638 (2011)] 2. Vaitkienė, Paulina; Skiriutė, Daina; Skauminas, Kęstutis; Tamašauskas, Arimantas. Associations between TFPI-2 methylation and poor prognosis in glioblastomas // Medicina. Kaunas : Lietuvos sveikatos mokslų universitetas. ISSN 1010-660X. 2012, t. 48, Nr. 7, p. 345-349 : pav, lent. <http://medicina.kmu.lt/1207/1207-03e.pdf>. [Science Citation Index Expanded (Web of Science); MEDLINE; Index Copernicus; DOAJ]. [Citav. rod.: 0,423 (2011)] 3. Skiriutė, Daina; Vaitkienė, Paulina; Šaferis, Viktoras; Ašmonienė, Virginija; Skauminas, Kęstutis; Deltuva, Vytenis Pranas; Tamašauskas, Arimantas. MGMT, GATA6, CD81, DR4, and CASP8 gene promoter methylation in glioblastoma // BMC cancer [electronic resource]. London : BioMed Central Ltd. (Research article). ISSN 1471-2407. 2012, vol. 12, 1 skelb. (218). <http://www.ncbi.nlm.nih.gov/pubmed/22672670>. [Science Citation Index Expanded (Web of Science); MEDLINE; CAS; SCOPUS; Google Scholar; EMBASE]. [Citav. rod.: 3,011 (2011)] 4. Vaitkienė, Paulina; Skiriutė, Daina; Mueller, Wolf. DNA methylation and gene expression of COX7A1, SPINT1 and KRT81 in glioblastoma and brain tissue // Biologija = Biology. Vilnius : Lietuvos mokslų akademijos leidykla. ISSN 1392-0146. 2012, t. 58, Nr. 2, p. 71-78 : pav, lent. [ISI Master Journal List; CABI Publishing; USDA Database; VINITI; EBSCO]. 129 Other publications 1. Steponaitis, Giedrius; Skiriutė, Daina; Vaitkienė, Paulina; Tamašauskas, Arimantas. Activity of MGMT gene in Glioblastoma as a predictive marker for treatment with alkylating agents. Biomedical engineering - 2012: Proceedings of international conference: 25, 26 October 2012: 90 metų KTU/Anniversary of KTU - 90 / Kaunas University of Technology. [Lithuanian society for biomedical engineering; Org. committee: A. Lukoševičius, A. Kriščiukaitis]. Kaunas: Technologija. ISSN 2029-3380. 2012, p. 5255 2. Vaitkienė Grigaitė, Paulina; Skiriutė, Daina; Ašmonienė, Virginija; Tamašauskas, Arimantas. DNA methylation impact on clinical outcome of glioblastoma patients. Biomedical engineering - 2011: Proceedings of International Conference: 27, 28 October 2011 / Kaunas University of Technology; [Org. committee: A. Lukoševičius, A. Kriščiukaitis]. Kaunas: Technologija. ISSN 20293380. 2011, p. 201-204 3. Vaitkienė, Paulina; Skiriutė, Daina; Skauminas, Kęstutis; Mueller, Wolf. Novel discovered targets of aberrant DNA methylation in glioblastomas. Biomedical engineering: Proceedings of International Conference: 2007 m. spalio 25, 26 d. / Kauno technologijos universitetas. Kaunas: Technologija, 2007. ISBN 9789955253679. p. 155-157 4. Vaitkienė, Paulina; Skiriutė, Daina; Steponaitis, Giedrius; Tamašauskas, Arimantas. Aberrant promoter DNA methylation in glioblastoma: impact on patient outcome. 5th Baltic congress of genetics: Kaunas, Lithuania 19-22 October, 2012. Kaunas, 2012. p. 64 (abstracts, poster) 5. Vaitkienė Grigaitė, Paulina; Skiriutė, Daina; Steponaitis, Giedrius; Ašmonienė, Virginija; Tamašauskas, Arimantas. Concomitant promoter methylation of multiple genes in glioblastoma. Lietuvos biochemikų draugijos XII konferencija "Biochemijos studijoms Lietuvoje - 50 metų": 2012 m. birželio mėn. 28-30 d, Tolieja (Molėtų raj.) / Lietuvos biochemikų draugija; [Org. komitetas: R. Daugelavičius (pirm.), ir kt.]. Kaunas: Vytauto Didžiojo universiteto leidykla, 2012. (Abstract, poster) p. 71, no. PS49, 67. 6. Vaitkienė Grigaitė, Paulina; Skiriutė, Daina; Mueller, Wolf. DNA methylation and gene expression of COX7A1, SPINT1 and KRT81 in glioblastoma and brain tissue. The Vital nature sign: 6th International scientific conference: Abstract book, 1st-4th June, 2012 130 / Vytautas Magnus University. Faculty of natural sciences. Kaunas, 2012. p. 34. And poster 7. Steponaitis, Giedrius; Skiriutė, Daina; Vaitkienė Grigaitė, Paulina; Tamašauskas, Arimantas. NDRG2 Promoter methylation status and gene expression level in benign and malignant brain tumors / Giedrius Steponaitis, Daina Skiriutė, Paulina Vaitkienė, Arimantas Tamašauskas // The Vital nature sign: 6th International scientific conference: Abstract book, 1st-4th June, 2012 / Vytautas Magnus University. Faculty of natural sciences. Kaunas, 2012. p. 25. And poster 8. Vaitkienė, Paulina; Skiriutė, Daina; Ašmonienė, Virginija; Steponaitis, Giedrius; Skauminas, Kęstutis; Kazlauskas, Arūnas; Tamašauskas, Arimantas. Promoter methylation of AREG, MLH1, HOXA11, NDRG2, NPTX2 and TES genes in glioblastoma. CSC Symposium Epigenetic Regulation: From Mechanism to Intervention: London 20–22 June 2012: These Abstracts / MRC Medical Research Council. Imperial College London; Scientific Organisers: Naill Dillon, Jesus Gil, Ana Pombo, Tim Aitman. Administration: Odeana Waldron, Tathiana Santana. London: Imperial College London, 2012. (Posters.). no. 115. 9. Vaitkienė Grigaitė, Paulina; Mueller, Wolf; Skauminas, Kęstutis. Gene regulation by methylation in glioblastomas. International Translational Neuroscience Conference: Celebrating the 60th Anniversary of Neurosurgery in Lithuania and establishing the Institute of Neurosciences of Lithuanian University of Health Sciences: Abstract book: 16-17 June 2011, Kaunas, Lithuania / [Institute for Biomedical Research, Lithuanian University of Health Sciences. Lithuanian Society of Skull Base Surgeons]. Kaunas: Published by the PI "Sorre", 2011. (Poster presentations.). ISBN 978-609-95133-6-2. p. 88, no. 71. 10. Skiriutė, Daina; Steponaitis, Giedrius; Vaitkienė Grigaitė, Paulina; Ašmonienė, Virginija; Tamašauskas, Arimantas. Investigation of promoter methylation of MGMT and GATA6 genes in Lithuanian glioblastoma patient’s tumor tissue. International Translational Neuroscience Conference: Celebrating the 60th Anniversary of Neurosurgery in Lithuania and establishing the Institute of Neurosciences of Lithuanian University of Health Sciences: Abstract book: 16-17 June 2011, Kaunas, Lithuania / [Institute for Biomedical Research, Lithuanian University of Health Sciences. Lithuanian Society of Skull Base Surgeons]. Kaunas: Published by the PI 131 "Sorre", 2011. (Poster presentations.). ISBN 978-609-95133-6-2. p. 42, no. 25. 11. Skiriutė, Daina; Vaitkienė Grigaitė, Paulina; Steponaitis, Giedrius; Ašmonienė, Virginija; Tamašauskas, Arimantas. MGMT, GATA6, CD81, DR4, CASP8 and NPTX2 gene promoter methylation study in glioblastomas. 22nd Wilhelm Bernhard Workshop: Program and Abstracts: August 25-29, 2011, Riga - Latvia / President: Nikolajs Sjakste; Universty of Latvia. Latvian Institute of Organic Synthesis. Riga, 2011. (Poster Session II.). p. 114, no. P44. 12. Ašmonienė, Virginija; Skiriutė, Daina; Vaitkienė Grigaitė, Paulina; Gudinavičienė, Inga; Tamašauskas, Šarūnas; Skauminas, Kęstutis; Deltuva, Vytenis Pranas; Tamašauskas, Arimantas. Primitive neuroectodermal tumor (pnet) located in third and fourth ventricles and frontal lobe: case report of a 51-years-old woman. European journal of human genetics - ESHG: European Human Genetics Conference 2009 : Vienna, Austria, May 23-26, 2009 : Abstracts. London: Nature Publishing Group. (Cancer genetics.). ISSN 10184813. 2009, vol. 17, suppl. 2, May, p. 181-182, no. P06.071 [Indėlis: 0,125] 13. Vaitkienė Grigaitė, Paulina; Mueller, Wolf; Laß, Ulrike; Skauminas, Kęstutis; Deltuva, Vytenis Pranas; Skiriutė, Daina; Tamašauskas, Arimantas; Deimling, Andreas. Frequent epigenetic silencing of NPTXII and AREG in glioblastomas. Second Nordic-Baltic postgraduate summer-school in cellular bioenergetics „Mitochondria in neurodegenerative and metabolic diseases: Tartu, June 10-15, 2007. Tartu, 2007. p. 34, no. 2. 14. Vaitkienė Grigaitė, Paulina; Mueller, Wolf; Laß, Ulrike; Deimling von, Andreas. Frequent epigenetic silencing of NPTXII and AREG in glioblastomas. Acta Neuropathologica: Joint Meeting of the German Society of Neuropathology and Neuroanatomy (DGNN) and the Polish Association of Neuropathologists with International Participation in Conjunction with the 52nd Annual Meeting of the German Society of Neuropathology and Neuroanatomy (DGNN) : Greifswald, Germany, September 05–08, 2007 / President: R. W. Warzok. Berlin: Springer Verlag. (Abstracts.). ISSN 0001-6322. 2007, vol. 114, no. 3, p. 329, no. 72. 15. Vaitkienė Grigaitė, Paulina; Mueller, Wolf; Laß, Ulrike; Deimling von, Andreas. Frequent epigenetic silencing of NPTXII and AREG in glioblastomas. The Baltic Summer School 2007: Inflammation: a key to common complex diseases: 2-13 September 2007, Lund, Sweden / Medical Inflammation Research, Department of 132 Experimental Medical Science Medical Faculty, Lund University. Lund: Medical Inflammation Research, Department of Experimental Medical Science, Lund University, [2007]. (Posters.). no. P41. 16. Vaitkienė Grigaitė, Paulina; Mueller, Wolf; Laß, Ulrike; Ehrlich, Matthew; Louis, David N.; Deimling, Andreas. Frequent epigenetic silencing of NPTXII in glioblastomas. Brain Tumor 2006: Program and Abstracts (Orals and Posters): December 7/8 2006, Berlin Berlin: HELIOS Kliniken, 2007. (Posters.). p. 19-20, no. 22. 133 ACKNOWLEDGMENTS Thank you so much for all that every single one of you did. This study would not have been possible without the contribution of many competent and kind persons. Therefore, I would like to acknowledge: My academic supervisor Prof. Dr. Dainius H. Pauža for accepting me as a doctoral student. His support and supervision throughout the years. My academic co-supervisor Prof. Habil. Dr. Arimantas Tamašauskas for providing me an attractive research project, large resources of patient material, continuous scientific support, encouragement and faith in my skills. I would like to thank the members of Laboratory of Neurooncology and Genetics of Neuroscience Institute (Lithuanian University of Health Sciences, Kaunas, Lithuania) for creating a friendly and enjoyable research environment. Dr. D. Skiriūtė for her supervision, introduction into the genetics of complex diseases. Her scientific supervision, support, encouragement provided the basis of this thesis. All clinicians team of the Kaunas University of Medicine Hospital, Department of Neurosurgery, for their expert help. Patients and their families, who agreed on the use of glioblastoma samples for academic research, for their cooperation. I also thank the technical staff at the Institute for Neuropathology, Charité Humboldt University (Berlin, Germany) for their expert help, pleasant and warm atmosphere. And finally, but not least, thanks goes to my whole family, who have been an important and indispensable source of spiritual support. 134 APPENDIX NPTX2 NPTX2gene promoter region with CpG islands is located chr7:97890357-97892510 Its sequence is as follows: TTACAAGGACCGGGCACTGTTGGACCACGTGGCTCCATCATGATGACTCCAGTA GATGTCACCCCGCCCCTGAGCTCAGGTCTTGCTGAATAAGGTCACCGCCCAGGG GGCAGTCGATGAACACGCGCGCGAGGGCTCTGCGAGTGGCCTCGTGACTTTGT CCCTAACTCCGGGTGTCCCCTCCTTCCCATCAGCGTCCGGCGCCTGGTCCTGGT CCCGGTCCCCGAGGCCCCCGGGATTCTTCCCGAGCGTTTTCCGAGTTGGCGCGG GGGGTGGAGGCGGGGCCATGGAGCGCGTCCCGGGGACCGTTGCATCCGGAGGC GGCCGTCGTGCGGCTCCTTCCCGCCTCGAGAGTGAGGTGGCCGGGCCTTGACG AGAAGGCCCACGCCTGCCGCGGGGGTGGCTCGCGATGGCAGTCGGGGTTCGAG TCCCGCCTGGGGGGCTGCTCCTGCTGGAGAAAACGCCTCCCTGAGGGCGGCGG CAAACGCGCAGCGAGGCCCCGTGCCGCGCCAGAAGCCACCCTGAGAAAGGGG CACCGGGACACCGAGGGGTTCCCACTTTCTCCTCAGCCTGTGACGCCCGCGTCC TCGGGTGGGTTCGAGGGGCGCCTGGGCACGGCCAGCCGAGGCTCTCGAGAGCC CCAGTGTCGTTTTCCACCTCAGGCCTCCTTTCCTGAGGCAGAGCCCGGGACCT CGCGCTCTCGCCTCAGGCTCCGGCCCACGCTCCCGCCCGGCCGCCAGGCGCGCA ACGGAAAGCGCCCCCGCCCCGCCCCGCTCCGCCCACTGCGTGACGCGCACCCG GCCGAGCCAATCAGAGCTCGTGGCGCGCGCCCCACACGCCGGCCCCCTCCGCC CCTCAGCTTAAGAAAGGGCGCGCGGACCCGGCAGGCCAGAGTGCCGAGCAGC GCGGTGGGTGCGGCTGTGAGACGGCAGGAGACTTCTGCCCCGCGGTGCACGCG ACCCTCGAGACGACAGCGCGGCTACTGCCAGCAGCGAAGGCGCCTCCCGCGGA GCGCCCCGACGGCGCCCGCTCGCCCATGCCGAGCTGAGCGCGGCAGCGGCGGC GGGATGCTGGCGCTGCTGGCCGCCAGCGTGGCGCTCGCCGTGGCCGCTGGGGC CCAGGACAGCCCGGCGCCCGGTAGCCGCTTCGTGTGCACGGCACTGCCCCCAG AGGCGGTGCACGCCGGCTGCCCGCTGCCCGCGATGCCCATGCAGGGCGGCGCG CAGAGTCCCGAGGAGGAGCTGAGGGCCGCGGTGCTGCAGCTGCGCGAGACCGT CGTGCAGCAGAAGGAGACGCTGGGCGCGCAGCGCGAGGCCATCCGCGAGCTC ACGGGCAAGCTAGCGCGCTGCGAGGGGCTGGCGGGCGGCAAGGCGCGCGGCG CGGGGGCCACGGGCAAGGACACTATGGGCGACCTGCCGCGGGACCCCGGCCAC GTCGTGGAGCAGCTCAGCCGCTCGCTGCAGACCCTCAAGGACCGCCTGGAGAG CCTCGAGGTAGCGGCCCGCGGGGAGCGCGGGGGACCTGGAATGGGGACGCTCC CGAGTCGGGGGCGGAAGACTCGGGAGGATGGGGAAAGGGGGCCTGGCCCTGG GGAGGGTGTGATCGTCCGTGGGGGTGAGCTGGACTTGAGGGTGAAAGGCGGGG ATCTAGATCCTGCTCGGGAACTCCCCTGCGTGGTATCCCTTCCCACACCGCTGC TCTTGCTGGAAGGAAACGTTTAAATTCCACCCCCGCGCGTCGGGACTGCCAGCG GGATCCGCCGAGCACTTCCCGAGGTCCGGGCTAGCGAACCCAGACGGCCAAGC CGCGGGCGCCAAATACCCGGGGACGCGGTAGCCTCTATCCTCTTGCAAATCTCC AAATCTCCGCGAGCCGGGATGCGCTCCCGCAGGCCGTGCGGGTTGCTGCGAGG CTGCCGCGGGAGGTCAGCTGCCCCGGGCATGTGGTGTTTCTTTCTTTAAATCAG GTCAGGCTGAGCTAGAGGGGGCGGAAGTCTGGGGGCGGAGGTCGCAGGGAGG CGAGCGCAGGGGGTCACCGGCGACCCCAGAGCGCGCATTAGAGGGGCAACCTC 135 TGGTTAACTTCAAAGACTCCCGTGTCTGCAAGTTGGACCATTTTAAGGTGCTCT TCCGGATTGCTTCAGGGGAAGGGACCTC Underlined marker: Bisulfite sequencing primers AREG Bold: Underscore: CpG-island, 454 bp exon 1, 271 bp TTTCAGAGTCAGATGAAGAATTCATATCCACCTGGCTTTGAACATTATCGGCTG TGAGATGGTGTAGGTAAAATTTTAAGTGCATAATTTGGCAATAATAAATCATCA ATAAATATTAATGTTGATGAGGCCCCTGGGCCACATAAAGAAATAGGGAGTGA GGGGATTTGAAATTCTGGCCACTTCACAGAAATGGGTGGGAAGGGGCTCTTGA TTGAGATAGAAGCCCATCCTACATGAAGCAATTCCTCATTGAGTTCTCTCGTCC TTTATCCTTGTTGGAAACATCAGGCAAAGTCACTCTTGGTCTTAAAGTACTTTTA CATCTAAATACGGAACTCTTCTATTTAATCCCTGTCTGTTGTAGATGTTAAGTAT ACAAAGAGGTTGTCAGAGTTTGAAACATCTGGACTTCTGTCAGGTACTAGCTCC GGAACTCCAGTCCTGCTCGCCCTCAAAAACGGCTTGCAGCTAGAGGTTTAAGTT CCACTTCCTCTCAGCGAATCCTTACGCACGAGGGAGGCGGGGCGTGTGTCC TCCGCGCGTGGTTTTCGGGTAGCACCTTCTGGGGCGCCGCCTGCCTCCAC CCACGGCCGGGCCTTGACGTCATGGGCTGCGGCCCCCTCCCGGCTGAGCC TATAAAGCGGCAGGTGCGCGCCGCCCTACAGACGTTCGCACACCTGGGTG CCAGCGCCCCAGAGGTCCCGGGACAGCCCGAGGCGCCGCGCCCGCCGCC CCGAGCTCCCCAAGCCTTCGAGAGCGGCGCACACTCCCGGTCTCCACTCG CTCTTCCAACACCCGCTCGTTTTGGCGGCAGCTCGTGTCCCAGAGACCGA GTTGCCCCAGAGACCGAGACGCCGCCGCTGCGAAGGACCAATGAGAGCCC CGCTGCTACCGCCGGCGCCGGTGGTGCTGTCGCTCTTGATACTCGGCTCA GGTGAGGATTCACCGGCGCTGAACTGCTGGGCTCTCCTCCCATGGCAGGTTTT GCCTCTTGAGTTTGGCTCTTGCTTCTCTAAAAATCGCCCTTTAGTTCCCCCGACC CTGCGCCGCAGGAGGTGATCACTGTAGCTCCCTCCCTAGGAGGAAAAGAGAAG GGCAGATGTCTCAAACGTCCAGGACAAGGTGTCCCAGAGAATCGCTGTACTTTT TCTTTAGTTCAGCTGAGAAGACGGGAGAGATCATTTGAACCTAACCCTTCGTTT CACAGATTAAAAGATTAAAAAGACCAAGATCTAGAGAGGTGAAGTGATTTCCG CAGATATTGGACCCAGCAGTGTTTGCTGCCTCCTGCCTAATTCTCCATCTCTGTC TAGTGATTTCTTCTTTACCTTCCTAGAGGGACTTAGAGTATCCCGGCGAGAGGT TAAAAGCACCACTCTGATGCTGTTTTATTGAGACCTAAGAAAAAGCAAGATAA AGCAGTGGCTTAACTCTGGGCCCCAAATCTTACT SPINT Bold: CpG-island, 1491 bp Underscore: exon 1; 139 bp Underscore: exon 2; 540 bp Underlined marker: Bisulfite sequencing primers GGATGTTGCAGTTCAGCAAAATCCAAGGGTTGGCCACAGGATCTGCTAGGCAA AGCTGCTCTGGATGAGAAACTGGGAAGAATGTTCCCATTGGTTTGGGGGTTGTT AAGCCATCAGGAAAAGGTTTGGAATCTGTATTTTAAGAGAGCATGCTATGTGA 136 TTCCTTCCTCCAGCCCTGCCTTCATCTGTCTTTCACCTGTTCAGACCCTTGCCGT AGAGAATGGTCTCTTAAGAGTTTTTTACTCAGGAGGAGCAATGAGCTTGGCTTC AAGGAGTCCACTGTTTCGTGAAGGGCATTAACTCAGCTACTTGGTGAACTGGA ACCTCCAGGCTTCCAAGGATGACATTTCCCTGGAAGAAAGTAGCCAGCAGCGT CCCCTTTGTGAAAGAGAGGAAGCCTGGGCTCCAGGGAAGGGGGCAACAGTCCA ATGAGCCATAGCAGGGAATGGACCGAACTGTACATACAAGATGAGGAGGGGC CTCGAACAACAGGGAAGCCCCGTGGAGCACGCAGGGGAGCCTGGACGCGG GGAATCGCCTCCGTTTCTCACCCTGACAGCAGGGAGGGCTTCCCGGAGCT GACAGTGAGGCAGACATCCCGGATCCCTGTTATCACAGGTGCAGCCACGC AACTTTTGGGTTACCTTCCCGCCACGAAATGGAAATGATTTCCTGAGGGCC GGAAAGATCATTTGCATCGGGCCTCAGGTCTGGGGCTGGGAAAGCAGGCC CCGGCCGGAAAGGAAGACTGGAGGCCGCCGCCCCAGAGGCCGGACCCTC GCGTCCCGGCCGCATCCCCTCCCGGCTGGGTATGCGCTTCGGGCGCGGGG CTCCCTCGCGAGGCTGGACTGGGTGGAGCCCGAGAGCCCCTCCGGGGAG CTGGCTGCCGGGGAGGCACCGGGGCTGCGGGGCCGCGGGGCGGGCCGGG CCTGGAGCTCCGCGCTAGAGGCGGCCGCAGCGCACCGGGCGTGGGCGGG GCCGGCAGCGCAAGGGTGAATGTCGGCCCCGCCCCGACGGCGCTGACTCA GTTTCACCAGAAACCAGGGGGAGAAGGCGGCCGAGCCCCAGCTCTCCGAG CACCGGGTCGGAAGCCGCGACCCGAGCCGCGCAGGAAGCTGGGACCGGA ACCTCGGCGGACCCGGCCCCACCCAACTCACCTGCGCAGGTAACCCGGGC CCCCGCGCGCAAGGCCGAGGCGCAGGCGGAGCGGGCTGGAGCATGTCCG GCCCTTTGTTCTGCGCTCGTGCGTGTGTCCGGGTACTTGAGCTCCCTAGCG GTCCGCCTGTCCGTCTGTCTGGCTGCCGGGTCCCTGGAGGGCGCGTGGGA GGGCCGGGCCCGTGCGCCCTCTTCGATCCTGGGGTGCTCCGGTCCCTCCT CCCCGCCACTTCCTCCCGGCCGGCCCGCCTCCTCCAAAGTCTCCCGGGCT GATCAGGTGTGTCTCCTCCTCTGTCCCCTCCCTTCTTCTCAGGTCACCAGC ACCCTCGGAACCCAGAGGCCCGCGCTCTGAAGGTGACCCCCCTGGGGAGG AAGGCGATGGCCCCTGCGAGGACGATGGCCCGCGCCCGCCTCGCCCCGG CCGGCATCCCTGCCGTCGCCTTGTGGCTTCTGTGCACGCTCGGCCTCCAG GGCACCCAGGCCGGGCCACCGCCCGCGCCCCCTGGGCTGCCCGCGGGAG CCGACTGCCTGAACAGCTTTACCGCCGGGGTGCCTGGCTTCGTGCTGGAC ACCAACGCCTCGGTCAGCAACGGAGCTACCTTCCTGGAGTCCCCCACCGT GCGCCGGGGCTGGGACTGCGTGCGCGCCTGCTGCACCACCCAGAACTGCA ACTTGGCGCTAGTGGAGCTGCAGCCCGACCGCGGGGAGGACGCCATCGCC GCCTGCTTCCTCATCAACTGCCTCTACGAGCAGAACTTCGTGTGCAAGTTC GCGCCCAGGGAGGGCTTCATCAACTACCTCACGAGGGAAGTGTACCGCTC CTACCGCCAGCTGCGGACCCAGGGCTTTGGAGGTGAGGAGGGTGCCAAGATG GATGGGTTTGGAGAGACTCAAAAAAGGGACTGGTTATGGGGTCCTAGGGGAGA CGAACATCAGAGAGGAGTTAAGGAGAGTTCACTTTTTTTTTTCTTTTTCTTTCTT TTTTTTTGCGACGGAGTCTCGCTCTTGTCGCCCAGGCTGGAGTGCAATGGCGCA ATCTCTGGTTGCTCACTGCAACCTCCGTCTTCCACGTTCAAGTGATTCTTCTGTC TCAGCCTCCGGAGTAGCTGGAACTACAGGCACCCGCTACCAGACCCGGCTAAT TTTTTTTTTTTTTTTTTTTTTTTTTTTTTTGGTACAGACGGGGTTTCACTATGTTGG CCAGGCTGGTCTTGAACTTCTGAACTCAGGTGATCCACCCACCTTGGCCTCCCA AAGTGCTGGGATTATTTCTACCTGAAGCCCTCCTTAGAAGTAGGCAGTACAGCC GAATCAAGCCTGGTGATCAATGGGGTGA 137 COX7A1 Bold: CpG-island, Underlined marker: Bisulfite sequencing primers ataccgttttactcccaaagactttagcttgtgtttcctaaaaacaaggtcatttaatgtctcaacaaatctctgagaaagatcctgcctta gccctcacGTTACAGATACGGAAACAGGCTCGGAGGTGCAATCTCTGGCTGTG TAGACAGAACAGGGAGTTGGAACACGGCCTTaTGCGTTTCCTCACTCTCAC ATACCAGCGGGCAGGGAGAGATCCGGGGAGGAGTTGCACTTGCCCCCGG CGGAAATCTCCAAAAATCTGCAAAAATGTATTCCCTGGTACCGCTTTGGGC TCCGGTGGGACCAAGCGAAATTCCGAGACCGAAACGGATGCGCGCTGCGG CCCAGGGTGCGGGTCTGGACCGCCTCCTCCGTGGTGGACGAATCCGAGGA GCAGGACTCCCCGCAACCCAGCCCCAGCCCCAGCCCCAGCCCCGCCGCGT CCCCAGCTGTCACGCTGCGCGCAGCGGGTGGGGCCTGGGGTTCCTGGACA GAGGAGGACTACGCGTGTCCTTGGGCGGAGAAGGGAGGTGACTCCGGCG GAAGAGGACAAGGCAGAATGCAGGCCCTTCGGgtgaggcccccatgggctgggccggagc cccccacccagaaaaccccgccctaatacgcgactctccaagtgcccccgcatctagggccttggaacgtgagccaccttc agggcctgggactccccgctaaacagcctgggtcccttcccccacaatccactcttcggagacaactggggacccccaccc cggcccaaagccccgggtttgaacttgacatctcgggaacccttaacactggcctcacccgggccttctcaggcagcctgga cgctcccggggacctccattccggtccccacgttcccgggacctggtttcctccctttaatccaggaaaccctcccccaacaa aaatctcaggacttaggccccccagaacaaagtccttctgggtcctctcccttccaggaccctctaggcagcccggtcaccc ccaggccagactccagggcgagtcccctcccaacccagctggcagttcgcgggtaacagaatccaagctattaccctcctc gcctttgggacgctcgctagggatggggctgtccccacgtccaggcctcagagagcggggtcccacttcaggacacccctc caggtgcctccaatacccgcccacctcccaccccgcagGTGTCCCAGGCGCTGATCCGCTCCTTC AGCTCCACCGCCCGGAACCGCTTTCAGAACCGAGTGCGCGAGAAACAGAA GCTCTTCCAGgtggggggtcggggggggtggggatccgacgcgccagccgggagcgcgccgagccggggcag gcggggcgcgctctaaggagcagccagcacccctttctcatcagacacccccacatccagGAGGACAATGACA TCCCGTTGTACCTGAAGGGCGGCATCGTTGACAACATCCTGTACCGAGTG ACAATGACGCTGTGTCTGGGCGgtgagcgcagggcccgtctgggctgcgggggaggcggggctgga cccagagtaagaggtggctggtttctgggcaggactgaccaggatctgggttgggggttggcgtttaggaagggggtcgag tactgagctggagtcaggcctgcagggaggggagtaactagggtttgggctcttgtcccaggacgggggctgggttagtgg tggggctgaggcccagggaggagtcggggagaaagctcccagcccggtctagatcaaagccagggggctttacaatagg agtccaatctgggaagggagctggagtccgaactagagctggggtccaggctgggaaccagggagcctgaaggccttaa aagcctgggatgtccagggaggggattaggtctcattctgtctccaacgcttcccctttctagGCACTGTCTACA GCTTGTACTCCCTTGGCTGGGCCTCCTTCCCCAGGAATTAAGACCAAGAAG CCTGGGGGGCCTGAGAGACTTGAACAAGTGTCAATAAACGCTGGCCTCTGt Two fragments were sequenced: The first fragment is located from –301 to +8 bp and covers 27 CG dinucleotides. The second fragment is located from +14 iki +363 and covers 17 CG dinucleotides. KRTB81 CpG-island Underlined marker: Bisulfite sequencing primers ATCTGAGATGAATAGTAAGTGGTGCTTTGGAATTATTGTTAATTTTCCTGAATA AGAAAATAGTACTCTCCTTATTCTTAAGAAATTCCTGCTGATGTATTTAAGGGT GACATGCCATGCTTTCTGCCACTCAAATAATTGGGCAACTGTGAAAGCAAATAT 138 GGTATAATGTTAACTGTTGAATCGGGGTAAACAGGTGGAGCATCAATGCATGT GTGTTGTATTATTCCAATTTTCCTCAATATTTGAAAGCTTTCATCATAAAAAAGA TAAAAGACAAATATTTAATAATCTGCTGACTGTTCCCATGGTAGGAAAAGTGA GATGGTTTTCAGCTGCCAACTGAAAAATCTGGAGATGGGGAAATCCAGATTTG TGTTTCAGGAGCACTTTAGCCACAGCTGTGATGGAAATGAGAGATGGTATTTAT TAATGCCTCAGCCTTTCCTGAAACCCCTCTCCCCACATCACACTTAACACACAG GTCTCCTCCTCCCAGTTTCTCCCAGCACCTGGCCTCCCTTCTCTTCCCCTCGCCA GGCTCTACTCCTCCTTCTCGTCCTCTCCTCTGCCTTCCCCTTCCCTCCCCAATGCC TGCAGATGAAGGAATGCCCTGCTGGCAAGACACTTTGAAGATGAAACATGCTG ACTCCCCCAGAGCCCAAGACTGACATCTTTTACAAAGAAGAGGGTGCAGGCCA CTCCCCCATTAAAGCACATTCTGAGAGGCGTTAGACCCGGCTAACCACCCAAG CCCATAAAGCGCAAATTGCCCCAACATCATCTTCACAGCCAAGCCCCTTCAGAA TCTGCGCATAAATAGGGCTGCGGTGCCCTGAGGAGCACATTGGAGTTTCCATCA GGACTCCAGGTCCCCTATCCTGTCCTCTGCAACCCAAACGTCCAGGAGGATCAT GACCTGCGGATCAGGATTTGGTGGGCGCGCCTTCAGCTGCATCTCGGCCTGC GGGCCGCGGCCCGGCCGCTGCTGCATCACCGCCGCCCCCTACCGTGGCAT CTCCTGCTACCGCGGCCTCACCGGGGGCTTCGGCAGCCACAGCGTGTGCG GAGGCTTTCGGGCCGGCTCCTGCGGACGCAGCTTCGGCTACCGCTCCGGG GGCGTGTGCGGGCCCAGTCCCCCATGCATCACCACCGTGTCGGTCAACGA GAGCCTCCTCACGCCCCTCAACCTGGAGATCGACCCCAACGCGCAGTGCG TGAAGCAGGAGGAGAAGGAGCAGATCAAGTCCCTCAACAGCAGGTTCGCGGC CTTCATCGACAAGGTGGGTGTCCTGGATCACACCCTTCCTGAACCCCCACCACC TACACAGCCAGGGCCGGGCACTAAGGATGGAGTCAGAGGCACAAAGACCTCTG CCTGCCTGAGATCCCAGTCTGATGGGCGAGGCACACAGACACACAGACAGACA GACACATGCACAGACACACACACACAGGCACTCAAGATGCAGACTCAGCTGAC AGTTCAGATAGAGGGAGCTTGTGAAGGAAAAGTGTGGCCAGGCTGCTCTGGGG CACAAGCTAGACATCAGGGAGTTGGGGGAGGGGGGCATGTGACCACTGCTGCT GAACTCAGAGGGGTTTGGGCAGCCCCAGGGTGAGAGAGTAGGGTGCACTGACT TCCAGATGACACAGGGGAAGGGCTGGGACAGCTGAGCTCCCTTCCAGATGGCC TGTGACACCCTCTTCCTGGCACCATCCCCAGAAACCTCATCTGTGATCTGAACC TGAGTACTCTGCCTGTGTGGGTTGCTGATCACCTAGTTTCATAGTGAGGGACTG AGCCTCCACTTCTGTTCATCTGCTTGGGTTGGGGGGGGGGGGTCACTCAACTCC CCAGGGAGCAGGCACTGACACCACCCAGCCCTTGCTCCACTCCTGAGTGGGAA ATCTAGACACAGATCATGACCAGGAAATCCAGAAATTGTCTGTCTCCCACTGCC CACCTCTACTCAATCTCAGATAAGCTCCAGATTTGGGGGTTCCCATTAAAGCAC CTCCCCTGCGAGCCCCCAGAGCTGGCTTTGGGGCACCAGACACCATCTGCCCCA CCTTCCCACACCGAATGGCAGGTGCGCTTCCTGGAGCAGCAGAACAAACTGCT GGAGACAAAGCTGCAGTTCTACCAGAACCGCGAGTGTTGCCAGAGCAACCTGG AGCCCCTGTTTGAGGGCTACATCGAGACTCTGCGGCGGGA The first fragment from – 85 to +239. It covers 26 CG dinucleotides. The second fragment from +213 to +388. It covers 11 CG dinucleotides. 139