Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 53
1.
PLoS Genet ; 15(8): e1008086, 2019 08.
Article En | MEDLINE | ID: mdl-31412020

DNA methyltransferases are ubiquitous enzymes conserved in bacteria, plants and opisthokonta. These enzymes, which methylate cytosines, are involved in numerous biological processes, notably development. In mammals and higher plants, methylation patterns established and maintained by the cytosine DNA methyltransferases (DMTs) are essential to zygotic development. In fungi, some members of an extensively conserved fungal-specific DNA methyltransferase class are both mediators of the Repeat Induced Point mutation (RIP) genome defense system and key players of sexual reproduction. Yet, no DNA methyltransferase activity of these purified RID (RIP deficient) proteins could be detected in vitro. These observations led us to explore how RID-like DNA methyltransferase encoding genes would play a role during sexual development of fungi showing very little genomic DNA methylation, if any. To do so, we used the model ascomycete fungus Podospora anserina. We identified the PaRid gene, encoding a RID-like DNA methyltransferase and constructed knocked-out ΔPaRid defective mutants. Crosses involving P. anserina ΔPaRid mutants are sterile. Our results show that, although gametes are readily formed and fertilization occurs in a ΔPaRid background, sexual development is blocked just before the individualization of the dikaryotic cells leading to meiocytes. Complementation of ΔPaRid mutants with ectopic alleles of PaRid, including GFP-tagged, point-mutated and chimeric alleles, demonstrated that the catalytic motif of the putative PaRid methyltransferase is essential to ensure proper sexual development and that the expression of PaRid is spatially and temporally restricted. A transcriptomic analysis performed on mutant crosses revealed an overlap of the PaRid-controlled genetic network with the well-known mating-types gene developmental pathway common to an important group of fungi, the Pezizomycotina.


Bacterial Proteins/physiology , DNA Modification Methylases/physiology , Gene Regulatory Networks/genetics , Podospora/physiology , Cytosine/metabolism , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Gene Expression Profiling , Gene Knockdown Techniques , Genes, Mating Type, Fungal/genetics , Genome, Bacterial
2.
Nucleic Acids Res ; 46(19): e113, 2018 11 02.
Article En | MEDLINE | ID: mdl-29986052

Efficient DNA assembly is of great value in biological research and biotechnology. Type IIS restriction enzyme-based assembly systems allow assembly of multiple DNA fragments in a one-pot reaction. However, large DNA fragments can only be assembled by alternating use of two or more type IIS restriction enzymes in a multi-step approach. Here, we present MetClo, a DNA assembly method that uses only a single type IIS restriction enzyme for hierarchical DNA assembly. The method is based on in vivo methylation-mediated on/off switching of type IIS restriction enzyme recognition sites that overlap with site-specific methylase recognition sequences. We have developed practical MetClo systems for the type IIS enzymes BsaI, BpiI and LguI, and demonstrated hierarchical assembly of large DNA fragments up to 218 kb. The MetClo approach substantially reduces the need to remove internal restriction sites from components to be assembled. The use of a single type IIS enzyme throughout the different stages of DNA assembly allows novel and powerful design schemes for rapid large-scale hierarchical DNA assembly. The BsaI-based MetClo system is backward-compatible with component libraries of most of the existing type IIS restriction enzyme-based assembly systems, and has potential to become a standard for modular DNA assembly.


Cloning, Molecular/methods , DNA Modification Methylases/physiology , Deoxyribonucleases, Type II Site-Specific/physiology , Base Sequence , DNA Ligases/metabolism , DNA Restriction Enzymes/classification , DNA Restriction Enzymes/physiology , DNA, Recombinant/biosynthesis , DNA, Recombinant/genetics , Escherichia coli , Gene Library , Synthetic Biology/methods
3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(4): 387-394, 2016 05 25.
Article Zh | MEDLINE | ID: mdl-27868412

The occurrence and development of acute myeloid leukemia (AML) is not only related to gene mutations, but also influenced by abnormal epigenetic regulation, in which DNA methylation is one of the most important mechanisms. Abnormal DNA methylation may lead to the activation of oncogene and the inactivation of tumor suppressor gene, resulting in the occurrence of leukemia. The mutations of DNA methylation enzymes associated with AML may have certain characteristics. The AML with recurrent cytogenetic abnormalities is also related to abnormal methylation. Some fusion genes can alter DNA methylation status to participate in the pathogenesis of leukemia. In addition, chemotherapy drug resistance in patients with AML is associated with the change of gene methylation status. Considering the reversibility of the epigenetic modification, targeted methylation therapy has become a hotspot of AML research.


DNA Methylation/drug effects , DNA Methylation/genetics , DNA Methylation/physiology , DNA Modification Methylases/genetics , Leukemia, Myeloid, Acute/genetics , DNA Modification Methylases/physiology , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/pathology , Mutation/genetics
4.
PLoS One ; 11(11): e0165509, 2016.
Article En | MEDLINE | ID: mdl-27824946

The silencing of the tumor suppressor gene O-6-methylguanine-DNA methyltransferase (MGMT) by promoter methylation commonly occurs in human cancers. The relationship between MGMT promoter methylation and gastric cancer (GC) remains inconsistent. This study aimed to evaluate the potential value of MGMT promoter methylation in GC patients. Electronic databases were searched to identify eligible studies. The pooled odds ratio (OR) and the corresponding 95% confidence interval (95% CI) were used to evaluate the effects of MGMT methylation on GC risk and clinicopathological characteristics. In total, 31 eligible studies including 2988 GC patients and 2189 nonmalignant controls were involved in meta-analysis. In the pooled analysis, MGMT promoter methylation was significantly associated with GC risk (OR = 3.34, P < 0.001) and substantial heterogeneity (P < 0.001). Meta-regression and subgroup analyses based on the testing method, sample material and ethnicity failed to explain the sources of heterogeneity. Interestingly, MGMT methylation showed a trend associated with gender, and methylation is lower in males compared with females (OR = 0.76, 95% CI = 0.56-1.03). We did not find a significant association in relation to tumor types, clinical stage, age status or H. pylori status in cancer (all P > 0.1). MGMT promoter methylation may be correlated with the prognosis of GCs in disease free survival (DFS) or overall survival (OS) for univariate analysis. MGMT promoter methylation may play a crucial role in the carcinogenesis and prognosis of GC. MGMT methylation was not correlated with tumor types, clinical stage, age status, H. pylori status. However, the result of the association of MGMT methylation and gender should be considered with caution.


DNA Methylation , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Promoter Regions, Genetic/physiology , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/physiology , Female , Humans , Male , Sex Factors , Stomach Neoplasms/etiology
5.
Adv Exp Med Biol ; 853: 111-38, 2015.
Article En | MEDLINE | ID: mdl-25895710

Glioblastoma remains a tumor with a dismal prognosis because of failure of current treatment. Glioblastoma cells with stem cell (GSC) properties survive chemotherapy and give rise to tumor recurrences that invariably result in the death of the patients. Here we summarize the current knowledge on chemoresistance of malignant glioma with a strong focus on GSC. Chemoresistant GSC are the most likely cause of tumor recurrence, but it remains controversial if GSC and under which conditions GSC are more chemoresistant than non-GSC within the tumor. Regardless of this uncertainty, the chemoresistance varies and it is mainly mediated by intrinsic factors. O6-methyl-guanidine methyltransferase (MGMT) remains the most potent mediator of chemoresistance, but disturbed mismatch repair system and multidrug resistance proteins contribute substantially. However, the intrinsic resistance by MGMT expression is regulated by extrinsic factors like hypoxia increasing MGMT expression and thereby resistance to alkylating chemotherapy. The search of new biomarkers helping to predict the tumor response to chemotherapy is ongoing and will complement the already known markers like MGMT.


Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm , Glioma/drug therapy , Neoplastic Stem Cells/drug effects , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Mismatch Repair/genetics , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/genetics , Glioma/genetics , Glioma/pathology , Humans , Neoplastic Stem Cells/pathology , Temozolomide , Tumor Suppressor Proteins/physiology
6.
Eur J Neurosci ; 41(6): 773-86, 2015 Mar.
Article En | MEDLINE | ID: mdl-25639476

Epigenetic mechanisms are increasingly acknowledged as major players in memory formation. Specifically, DNA methylation is necessary for the formation of long-term memory in various brain regions, including the hippocampus (HPC); however, its role in the perirhinal cortex (PRh), a structure critical for object memory, has not been characterized. Moreover, the mnemonic effects of selective DNA methyltransferase (DNMT) inhibition have not yet been investigated systematically, despite distinct roles for de novo (DNMT3a, 3b) and maintenance (DNMT1) methyltransferases. Consequently, we assessed the effects of various DNMT inhibitors within the HPC and PRh of rats using the object-in-place paradigm, which requires both brain regions. The non-nucleoside DNA methyltransferase inhibitor RG-108 impaired long-term object-in-place memory in both regions. Furthermore, intracranial administration of Accell short-interference RNA sequences to inhibit the expression of individual DNMTs implicated DNMT3a and DNMT1 in the HPC and PRh effects, respectively. mRNA expression analyses revealed a complementary pattern of results, as only de novo DNMT3a and DNMT3b mRNA was upregulated in the HPC (dentate gyrus) following object-in-place learning, whereas DNMT1 mRNA was selectively upregulated in the PRh. These results reinforce the established functional double dissociation between the HPC and PRh and imply the operation of different epigenetic mechanisms in brain regions dedicated to long-term memory processing for different types of information.


Cerebral Cortex/physiology , DNA Modification Methylases/physiology , Hippocampus/physiology , Memory, Long-Term/physiology , Animals , Cerebral Cortex/drug effects , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methyltransferase 3A , DNA Modification Methylases/antagonists & inhibitors , Hippocampus/drug effects , Male , Memory, Long-Term/drug effects , Phthalimides/pharmacology , Rats , Rats, Long-Evans , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
7.
Actas dermo-sifiliogr. (Ed. impr.) ; 106(1): 17-28, ene.-feb. 2015. ilus, tab
Article Es | IBECS | ID: ibc-133274

La homeostasis de la piel, cuya regulación molecular es aún bastante desconocida, está íntimamente relacionada con la función de las células madre epidérmicas. El programa SkinModel-CM, auspiciado por la Comunidad de Madrid, reúne 5 grupos de investigación con el propósito de desarrollar nuevos modelos experimentales in vitro e in vivo para analizar la función de ADN metiltransferasa 1, la endoglina y la podoplanina en la actividad de las células madre epidérmicas y en la homeostasis y el cáncer cutáneos. Estos nuevos modelos comprenden tanto cultivos organotípicos 3 D, como ratones inmunodeficientes con la piel humanizada y ratones modificados genéticamente. Otro objetivo del programa es el uso de ratones con la piel humanizada como modelo para reconstruir enfermedades cutáneas, tales como el síndrome de Gorlin y el xeroderma pigmentoso, con el objeto de optimizar nuevos protocolos de intervención mediante la terapia fotodinámica (AU)


Homeostasis, whose regulation at the molecular level is still poorly understood, is intimately related to the functions of epidermal stem cells. Five research groups have been brought together to work on new in vitro and in vivo skin models through the SkinModel-CM program, under the auspices of the Spanish Autonomous Community of Madrid. This project aims to analyze the functions of DNA methyltransferase 1, endoglin, and podoplanin in epidermal stem cell activity, homeostasis, and skin cancer. These new models include 3-dimensional organotypic cultures, immunodeficient skin-humanized mice, and genetically modified mice. Another aim of the program is to use skin-humanized mice to model dermatoses such as Gorlin syndrome and xeroderma pigmentosum in order to optimize new protocols for photodynamic therapy (AU)


Humans , Homeostasis/physiology , Skin Diseases/physiopathology , DNA Modification Methylases/physiology , Stem Cells/physiology , Disease Models, Animal , Phototherapy , Hair Follicle/physiology , Models, Genetic , Bioengineering/methods
8.
Environ Sci Pollut Res Int ; 22(21): 16262-76, 2015 Nov.
Article En | MEDLINE | ID: mdl-25172464

Environmental epigenetics is a rapidly growing field which studies the effects of environmental factors such as nutrition, stress, and exposure to compounds on epigenetic gene regulation. Recent studies have shown that exposure to toxicants in vertebrates is associated with changes in DNA methylation, a major epigenetic mechanism affecting gene transcription. Zebra fish, a well-known model in toxicology and developmental biology, are emerging as a model species in environmental epigenetics despite their evolutionary distance to rodents and humans. In this review, recent insights in DNA methylation during zebra fish development are discussed and compared to mammalian models in order to evaluate zebra fish as a model to study the role of DNA methylation in environmental toxicology. Differences exist in DNA methylation reprogramming during early development, whereas in later developmental stages, tissue distribution of both 5-methylcytosine and 5-hydroxymethylcytosine seems more conserved between species, as well as basic DNA (de)methylation mechanisms. All DNA methyl transferases identified so far in mammals are present in zebra fish, as well as a number of major demethylation pathways. However, zebra fish appear to lack some methylation pathways present in mammals, such as parental imprinting. Several studies report effects on DNA methylation in zebra fish following exposure to environmental contaminants, such as arsenic, benzo[a]pyrene, and tris(1,3-dichloro-2-propyl)phosphate. Though more research is needed to examine heritable effects of contaminant exposure on DNA methylation, recent data suggests the usefulness of the zebra fish as a model in environmental epigenetics.


DNA Methylation , Environmental Pollutants/toxicity , Epigenesis, Genetic/drug effects , Animals , DNA Modification Methylases/physiology , Ecotoxicology , Gene Expression Regulation, Developmental , Genomic Imprinting , Humans , Zebrafish , Zebrafish Proteins/physiology
9.
J Biol Regul Homeost Agents ; 29(4): 853-8, 2015.
Article En | MEDLINE | ID: mdl-26753647

Glioma is a serious life-threatening disease, the pathogenesis of which remains to be investigated. The objective of the present investigation was to explore the expression and clinical significance of tumor suppressor gene (P53), O6-methylguanine-DNA methyltransferase (MGMT) and epidermal growth factor receptor (EGFR) in glioma. Immunohistochemical staining was applied to study the clinical characteristics of 40 samples from glioma patients, detect the expression of and analyse the relationship between P53, MGMT and EGFR and glioma. The results demonstrated that the positive expression rate of P53 was 47.5% in 40 cases of glioma samples, of which the expression of P53 in the high grade glioma was higher than that of the low grade samples (P < 0.05); the positive expression rate of MGMT was 37.5%, but there was no significant significance of MGMT expression between the high grade glioma and the low grade glioma (P >0.05); the positive expression rate of EGFR was 55%, of which the expression of EGFR of the high grade glioma was higher than that of the low grade glioma (P<0.05). There was no significant difference in the expressions of P53, MGMT and EGFR in the glioma patients of different ages, gender and with different tumor sizes. The expressions of P53 and MGMT were negatively correlated (P<0.05). The expressions of P53 and EGFR were positively correlated (P<0.05). In conclusion, P53, EGFR and MGMT could play a role in the occurrence, development and deterioration of glioma.


Brain Neoplasms/pathology , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , ErbB Receptors/physiology , Glioma/pathology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Adolescent , Adult , Aged , Brain Neoplasms/chemistry , Brain Neoplasms/etiology , Child , DNA Modification Methylases/analysis , DNA Repair Enzymes/analysis , ErbB Receptors/analysis , Female , Glioma/chemistry , Glioma/etiology , Humans , Male , Middle Aged , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Proteins/analysis
11.
New Phytol ; 203(1): 140-54, 2014 Jul.
Article En | MEDLINE | ID: mdl-24697753

Exine, the outermost layer of a pollen grain, has important roles in protecting microspore cytoplasm and determining species-specific interactions between pollen and stigma. The molecular mechanism underlying pollen exine formation, however, remains largely unknown. Here, we report the characterization of an Arabidopsis male-sterile mutant, efd, which exhibits male sterility in first-forming flowers. The Exine Formation Defect (EFD) gene is strongly expressed in microsporocytes, tetrads and the tapetum, and encodes a nuclear-localized de novo DNA methyltransferase. Detailed observations revealed that EFD is involved in both callose wall and primexine formation during microsporogenesis. Microspores in tetrads are not well separated in efd due to an abnormal callose wall. Its plasma membrane undulation appears normal, but primexine patterning is impaired. Primexine matrix establishment and sporopollenin accumulation at specific positions are disturbed, and thus exine formation is totally blocked in efd. We confirmed that EFD is required for pollen exine formation and male fertility via the regulation of callose wall and primexine formation. We also found that positional sporopollenin accumulation is not involved in regulating membrane undulation, but is related to the complete separation of tetrad microspores during primary exine patterning.


Arabidopsis Proteins/physiology , Arabidopsis/physiology , DNA Modification Methylases/physiology , Pollen/physiology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , DNA Methylation , DNA Modification Methylases/genetics , Gametogenesis, Plant/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Plant , Mutagenesis, Insertional , Plant Infertility/genetics
12.
J Clin Invest ; 124(1): 17-23, 2014 Jan.
Article En | MEDLINE | ID: mdl-24382385

The Cancer Genome Atlas (TCGA) Research Network is an ambitious multi-institutional consortium effort aimed at characterizing sequence, copy number, gene (mRNA) expression, microRNA expression, and DNA methylation alterations in 30 cancer types. TCGA data have become an extraordinary resource for basic, translational, and clinical researchers and have the potential to shape cancer diagnostic and treatment strategies. DNA methylation changes are integral to all aspects of cancer genomics and have been shown to have important associations with gene expression, sequence, and copy number changes. This Review highlights the knowledge gained from DNA methylation alterations in human cancers from TCGA.


DNA Methylation , Epigenesis, Genetic , Neoplasms/genetics , Animals , BRCA1 Protein/genetics , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Genome, Human , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasms/enzymology , Tumor Suppressor Proteins/physiology
13.
Neurobiol Learn Mem ; 107: 93-100, 2014 Jan.
Article En | MEDLINE | ID: mdl-24291571

We have previously shown that auditory Pavlovian fear conditioning is associated with an increase in DNA methyltransferase (DNMT) expression in the lateral amygdala (LA) and that intra-LA infusion or bath application of an inhibitor of DNMT activity impairs the consolidation of an auditory fear memory and long-term potentiation (LTP) at thalamic and cortical inputs to the LA, in vitro. In the present study, we use awake behaving neurophysiological techniques to examine the role of DNMT activity in memory-related neurophysiological changes accompanying fear memory consolidation and reconsolidation in the LA, in vivo. We show that auditory fear conditioning results in a training-related enhancement in the amplitude of short-latency auditory-evoked field potentials (AEFPs) in the LA. Intra-LA infusion of a DNMT inhibitor impairs both fear memory consolidation and, in parallel, the consolidation of training-related neural plasticity in the LA; that is, short-term memory (STM) and short-term training-related increases in AEFP amplitude in the LA are intact, while long-term memory (LTM) and long-term retention of training-related increases in AEFP amplitudes are impaired. In separate experiments, we show that intra-LA infusion of a DNMT inhibitor following retrieval of an auditory fear memory has no effect on post-retrieval STM or short-term retention of training-related changes in AEFP amplitude in the LA, but significantly impairs both post-retrieval LTM and long-term retention of AEFP amplitude changes in the LA. These findings are the first to demonstrate the necessity of DNMT activity in the consolidation and reconsolidation of memory-associated neural plasticity, in vivo.


Amygdala/enzymology , Amygdala/physiology , DNA Modification Methylases/physiology , Memory/physiology , Neuronal Plasticity/physiology , Acoustic Stimulation , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Fear/physiology , Male , Memory/drug effects , Mental Recall/drug effects , Mental Recall/physiology , Neuronal Plasticity/drug effects , Phthalimides/pharmacology , Rats , Rats, Sprague-Dawley , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
14.
Med Hypotheses ; 80(6): 728-31, 2013 Jun.
Article En | MEDLINE | ID: mdl-23541771

The isocitrate dehydrogenase 1 (IDH1) mutation occurs in high frequency in glioma and secondary glioblastoma (GBM). Mutated IDH1 produces the oncometabolite 2-hydroxyglutarate rather than α-ketoglutarate or isocitrate. The oncometabolite is considered to be the major cause of the association between the IDH1 mutation and gliomagenesis. On the other hand, the IDH1 mutation in GBM is associated with prolonged patient survival. This association is not well understood yet but IDH1 involvement in epigenetic silencing of O-6-methylguanine-DNA methyltransferase (MGMT), a DNA repair enzyme is considered to be an important mechanism. However, it was shown recently that the IDH1 mutation and MGMT silencing are independent prognostic factors. Here, we hypothesize that the IDH1 mutation reduces the capacity to produce NADPH and thus reduces the capacity to scavenge reactive oxygen species that are generated during irradiation and chemotherapy. IDH1 activity is responsible for two-thirds of the NADPH production capacity in normal brain, whereas the IDH1 mutation reduces this capacity by almost 40%. Therefore, we hypothesize that the reduced NADPH production capacity due to the IDH1 mutation renders GBM cells more vulnerable to irradiation and chemotherapy thus prolonging survival of the patients.


DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Glioma/genetics , Glioma/mortality , Isocitrate Dehydrogenase/genetics , NADP/biosynthesis , Tumor Suppressor Proteins/physiology , Chemoradiotherapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gene Silencing/physiology , Glioma/metabolism , Glioma/therapy , Humans , Isocitrate Dehydrogenase/metabolism , Models, Biological , Mutation/genetics , NADP/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/genetics
15.
Surg Today ; 43(12): 1355-64, 2013 Dec.
Article En | MEDLINE | ID: mdl-23291904

Epigenetic changes such as DNA methylation, histone modification, and loss of genome imprinting play a crucial role in esophageal squamous cell carcinogenesis, along with genomic and genetic alterations. DNA methylation is a fundamental epigenetic process that modulates gene expression. Cancer cells exhibit two types of alterations of DNA methylation: global DNA hypomethylation and site-specific CpG island promoter hypermethylation. In several types of human cancers, the methods of detecting an aberrant methylation status have been applied to clinical fields to stratify high-risk groups, detect early cancer, and predict clinical outcomes. Importantly, epigenetic changes, including alterations in DNA methylation, are reversible and can thus be targets for cancer therapy or chemoprevention. Therefore, a better understanding of the DNA methylation in esophageal squamous cell carcinoma (ESCC) is important for optimizing cancer therapy and chemoprevention. We herein summarize the current knowledge regarding alterations in DNA methylation and the clinical implications in ESCC.


Carcinoma, Squamous Cell/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Esophageal Neoplasms/genetics , Long Interspersed Nucleotide Elements/genetics , Adaptor Proteins, Signal Transducing , Carcinoma, Squamous Cell/therapy , CpG Islands/genetics , DNA Mismatch Repair/genetics , DNA Modification Methylases/physiology , DNA Repair/genetics , DNA Repair Enzymes/physiology , Esophageal Neoplasms/therapy , Gene Silencing , Genes, Tumor Suppressor , Humans , MicroRNAs , Molecular Targeted Therapy , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Nuclear Proteins , Tumor Suppressor Proteins/physiology
16.
J Biol Chem ; 287(40): 33116-21, 2012 Sep 28.
Article En | MEDLINE | ID: mdl-22898819

For cytosine (C) demethylation of vertebrate DNA, it is known that the TET proteins could convert 5-methyl C (5-mC) to 5-hydroxymethyl C (5-hmC). However, DNA dehydroxymethylase(s), or enzymes able to directly convert 5-hmC to C, have been elusive. We present in vitro evidence that the mammalian de novo DNA methyltransferases DNMT3A and DNMT3B, but not the maintenance enzyme DNMT1, are also redox-dependent DNA dehydroxymethylases. Significantly, intactness of the C methylation catalytic sites of these de novo enzymes is also required for their 5-hmC dehydroxymethylation activity. That DNMT3A and DNMT3B function bidirectionally both as DNA methyltransferases and as dehydroxymethylases raises intriguing and new questions regarding the structural and functional aspects of these enzymes and their regulatory roles in the dynamic modifications of the vertebrate genomes during development, carcinogenesis, and gene regulation.


DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Modification Methylases/physiology , Animals , Cell Nucleus/metabolism , Chromatography, Thin Layer/methods , DNA/chemistry , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methylation , DNA Methyltransferase 3A , DNA Modification Methylases/chemistry , Dioxygenases/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Models, Biological , Oxidation-Reduction , Plasmids/metabolism , DNA Methyltransferase 3B
18.
J Exp Med ; 208(4): 689-702, 2011 Apr 11.
Article En | MEDLINE | ID: mdl-21464220

Glioblastoma Multiforme (GBM), the most common and lethal primary human brain tumor, exhibits multiple molecular aberrations. We report that loss of the transcription factor GATA4, a negative regulator of normal astrocyte proliferation, is a driver in glioma formation and fulfills the hallmarks of a tumor suppressor gene (TSG). Although GATA4 was expressed in normal brain, loss of GATA4 was observed in 94/163 GBM operative samples and was a negative survival prognostic marker. GATA4 loss occurred through promoter hypermethylation or novel somatic mutations. Loss of GATA4 in normal human astrocytes promoted high-grade astrocytoma formation, in cooperation with other relevant genetic alterations such as activated Ras or loss of TP53. Loss of GATA4 with activated Ras in normal astrocytes promoted a progenitor-like phenotype, formation of neurospheres, and the ability to differentiate into astrocytes, neurons, and oligodendrocytes. Re-expression of GATA4 in human GBM cell lines, primary cultures, and brain tumor-initiating cells suppressed tumor growth in vitro and in vivo through direct activation of the cell cycle inhibitor P21(CIP1), independent of TP53. Re-expression of GATA4 also conferred sensitivity of GBM cells to temozolomide, a DNA alkylating agent currently used in GBM therapy. This sensitivity was independent of MGMT (O-6-methylguanine-DNA-methyltransferase), the DNA repair enzyme which is often implicated in temozolomide resistance. Instead, GATA4 reduced expression of APNG (alkylpurine-DNA-N-glycosylase), a DNA repair enzyme which is poorly characterized in GBM-mediated temozolomide resistance. Identification and validation of GATA4 as a TSG and its downstream targets in GBM may yield promising novel therapeutic strategies.


Brain Neoplasms/prevention & control , GATA4 Transcription Factor/physiology , Glioblastoma/prevention & control , Tumor Suppressor Proteins/physiology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Methylation , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , GATA4 Transcription Factor/genetics , Glioblastoma/pathology , Humans , Mice , Promoter Regions, Genetic , Temozolomide
19.
Brain Tumor Pathol ; 28(1): 25-31, 2011 Feb.
Article En | MEDLINE | ID: mdl-21249460

Recent reports have suggested an important clinical role for hypermethylation of the O(6)-methylguanine-DNA-methyltransferase (MGMT) promoter in patients with glioblastomas. Whether MGMT protein expression is correlated with promoter hypermethylation and patient outcomes, however, has not been elucidated. Here we describe a quantitative digital method for assessment of MGMT-specific immunostaining, and analyze the relationship between expression levels and methylation status of the MGMT promoter. We investigated 46 tumors from patients who received a diagnosis of glioblastoma or gliosarcoma. Immunohistochemistry with anti-MGMT antibody and methylation-specific PCR using bisulfite-modified tumor DNA were performed. The digital assessment method used image-analysis software to determine a digital MGMT staining index, and the results were compared with those obtained via conventional visual assessments. The digital staining index clearly correlated with the methylation status of MGMT promoter. In addition, the index correlated with our observational results when nuclear and cytoplasmic staining were assessed in three different fields. Our digital assessment method enabled us to assess uncertain immunopositive samples objectively and quantitatively, which is an important consideration when examining heterogeneous cellular staining. We expect that this method will be useful for assessment of heterogeneous staining with any antibodies.


Biomarkers, Tumor/analysis , Brain Neoplasms/diagnosis , DNA Modification Methylases/analysis , DNA Modification Methylases/physiology , DNA Repair Enzymes/analysis , DNA Repair Enzymes/physiology , Glioblastoma/diagnosis , Gliosarcoma/diagnosis , Immunohistochemistry/methods , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/physiology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Female , Glioblastoma/drug therapy , Gliosarcoma/drug therapy , Humans , Male , Methylation , Middle Aged
20.
Clin Transl Oncol ; 12(6): 401-9, 2010 Jun.
Article En | MEDLINE | ID: mdl-20534395

In this study, we reviewed epigenetic therapy of lymphomas using histone deacetylase inhibitors (HDACi), a promising new class of antineoplastic agents. Epigenetic therapy, a new therapeutic concept, consists of the use of HDACi and or DNA methyltransferase inhibitors (DNMTi). We conducted a comprehensive review of the literature for antitumour activity of HDACi and its mechanism of action. HDACi modify the expression of several genes related to cancer development, which can result in antineoplastic activity. To elucidate the benefits of HDACi in lymphoma treatment, we discuss the crucial interplay between BCL6, p53 and STAT3. Activated B-cell (ABC) diffuse large cell lymphoma (DLCL) is increasingly being recognised as an unfavourable and frequently therapy-refractory lymphoma. We discuss the fundamental causative role of the STAT3 oncogene in ABC type DLCL. STAT3 can be effectively suppressed by several HDACi, a promising treatment for this difficult subtype of DLCL. On the other hand, various HDACi can repress the germinal-centre B Cell (GCB) type DLCL by virtue of their inhibition of the BCL6 oncogene, usually expressed in this particular subtype. We summarise the results of recent clinical trials with HDACi such as romidepsin, panobinostat, MGCD-0103, entinostat, curcumin, JAK2 inhibitor TG101348, and valproic acid that have shown preliminary activity in recurrent and refractory lymphomas. The unique mechanism of action of HDACi makes them very attractive agents to pursue in combination. Several ongoing trials are already exploring HDACi combinations in various types of cancers. Their role in front-line management remains to be determined.


Antineoplastic Agents/therapeutic use , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Lymphoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , DNA Modification Methylases/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Lymphoma/genetics , Models, Biological , Signal Transduction/drug effects
...