Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Mol Med ; 24(17): 9898-9907, 2020 09.
Article in English | MEDLINE | ID: mdl-32693431

ABSTRACT

Congenital heart disease (CHD) with extracardiac malformations (EM) is the most common multiple malformation, resulting from the interaction between genetic abnormalities and environmental factors. Most studies have attributed the causes of CHD with EM to chromosomal abnormalities. However, multi-system dysplasia is usually caused by both genetic mutations and epigenetic dysregulation. The epigenetic mechanisms underlying the pathogenesis of CHD with EM remain unclear. In this study, we investigated the mechanisms of imprinting alterations, including those of the Small nuclear ribonucleoprotein polypeptide N (SNRPN), PLAG1 like zinc finger 1 (ZAC1) and inositol polyphosphate-5-phosphatase F (INPP5F) genes, in the pathogenesis of CHD with EM. The methylation levels of SNRPN, ZAC1, and INPP5F genes were analysed by the MassARRAY platform in 24 children with CHD with EM and 20 healthy controls. The expression levels of these genes were detected by real-time polymerase chain reaction (PCR). The correlation between methylation regulation and gene expression was confirmed using 5-azacytidine (5-Aza) treated cells. The methylation levels of SNRPN and ZAC1 genes were significantly increased in CHD with EM, while that of INPP5F was decreased. The methylation alterations of these genes were negatively correlated with expression. Risk analysis showed that abnormal hypermethylation of SNRPN and ZAC1 resulted in 5.545 and 7.438 times higher risks of CHD with EM, respectively, and the abnormal hypomethylation of INPP5F was 8.38 times higher than that of the control group. We concluded that abnormally high methylation levels of SNRPN and ZAC1 and decreased levels of INPP5F imply an increased risk of CHD with EM by altering their gene functions. This study provides evidence of imprinted regulation in the pathogenesis of multiple malformations.


Subject(s)
Cell Cycle Proteins/genetics , Heart Defects, Congenital/genetics , Inositol Polyphosphate 5-Phosphatases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , snRNP Core Proteins/genetics , Azacitidine/chemistry , Child , Child, Preschool , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Female , Gene Expression/genetics , Genomic Imprinting/genetics , Heart Defects, Congenital/epidemiology , Heart Defects, Congenital/pathology , Humans , Infant , Male
2.
Analyst ; 145(8): 3064-3072, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32141455

ABSTRACT

Sensitive and accurate determination of DNA methyltransferase (DNA Mtase) activity is highly pursued for understanding fundamental biological processes related to DNA methylation, clinical disease diagnosis and drug discovery. Herein, we propose a new electrochemical immuno-DNA sensing platform for DNA Mtase activity assay and inhibitor screening. After homogeneous DNA methylation by CpG methyltransferase (M.SssI Mtase), the methylated DNA can be specifically recruited onto an electrode via its immunological binding with the immobilized anti-5-methylcytosine antibody. The recruited methylated DNA was simultaneously used as a substrate to facilitate successive template-free DNA extension and enzyme catalysis for the dual-step signal amplification of DNA Mtase activity. The developed immuno-DNA sensing strategy effectively integrates solution-phase DNA methylation, surface affinity binding recognition, and successive template-free DNA extension and enzyme catalysis-based signal amplification, rendering a highly specific, sensitive and accurate assay of DNA Mtase activity. A low detection limit of 0.039 U mL-1 could be achieved with a high selectivity. It was also applied for efficient evaluation of various inhibitors. Current affinity recognition of the immobilized antibody with methylated DNA switches the sensing platform into a DNA operation interface, facilitating the opportunity for combining various DNA-based signal amplification strategies to improve the detection performance. It would be used as a general strategy for the analysis of DNA Mtase activity, inhibitors and more analytes, and is anticipated to show potential for applications in disease diagnosis and drug discovery.


Subject(s)
DNA-Cytosine Methylases/analysis , DNA/chemistry , Electrochemical Techniques/methods , Enzyme Assays/methods , Enzyme Inhibitors/chemistry , Immunoassay/methods , Animals , Antibodies, Immobilized/immunology , Azacitidine/chemistry , Biosensing Techniques/methods , DNA/immunology , DNA Nucleotidylexotransferase/chemistry , DNA-Cytosine Methylases/antagonists & inhibitors , Decitabine/chemistry , Electrochemical Techniques/instrumentation , Electrodes , Limit of Detection , Mice
3.
Methods ; 156: 60-65, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30308313

ABSTRACT

Well over a hundred types of naturally occurring covalent modifications can be made to ribonucleotides in RNA molecules. Moreover, several types of such modifications are each known to be catalysed by multiple enzymes which largely appear to modify distinct sites within the cellular RNA. In order to aid functional investigations of such multi-enzyme RNA modification types in particular, it is important to determine which enzyme is responsible for catalysing modification at each site. Two methods, Aza-IP and methylation-iCLIP, were developed and used to map genome-wide locations of methyl-5-cytosine (m5C) RNA modifications inherently in an enzyme specific context. Though the methods are quite distinct, both rely on capturing catalytic intermediates of RNA m5C methyltransferases in a state where the cytosine undergoing methylation is covalently crosslinked to the enzyme. More recently the fundamental methylation-iCLIP principle has also been applied to map methyl-2-adenosine sites catalysed by the E. coli RlmN methylsynthase. Here I describe the ideas on which the two basic methods hinge, and summarise what has been achieved by them thus far. I also discuss whether and how such principles may be further exploited for profiling of other RNA modification types, such as methyl-5-uridine and pseudouridine.


Subject(s)
Escherichia coli Proteins/metabolism , Immunoprecipitation/methods , Methyltransferases/metabolism , Multienzyme Complexes/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/chemistry , Transcriptome , Animals , Azacitidine/chemistry , Azacitidine/metabolism , Biocatalysis , Cross-Linking Reagents/chemistry , Escherichia coli Proteins/genetics , Fluorouracil/chemistry , Fluorouracil/metabolism , Humans , Methylation , Methyltransferases/genetics , Multienzyme Complexes/genetics , Pseudouridine/chemistry , Pseudouridine/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uridine/analogs & derivatives , Uridine/chemistry , Uridine/metabolism
4.
G Ital Med Lav Ergon ; 42(2): 109-120, 2020 06.
Article in English | MEDLINE | ID: mdl-32614541

ABSTRACT

SUMMARY: Antineoplastic drugs are used to treat cancer, having their therapeutic effect by inhibiting the cell division process. Although cancer cells, due to their rapid growth, are more sensitive to the toxic effects of chemotherapeutic agents, healthy cells and tissues may also be damaged. Many studies show acute and chronic toxicity both in patients treated with chemotherapy and in exposed workers. In fact, exposure to these substances can also be linked to the formation of different types of secondary tumors. The International Agency on Research on Cancer (IARC) included some antineplastic drugs in Group 1 (carcinogenic to humans), in Group 2A (probable carcinogens for In recent years, many studies have evidenced the presence of antineoplastic drug contamination on work surfaces, materials and floors and based on these observations, international and national guidelines have been published to limit occupational exposure, with particular attention to procedures post-preparation of chemotherapy to limit as much as possible the accumulation of contaminated residues. The aim of the following study is to determine the effectiveness of the degradation of four antineoplastic drugs: 5-fluorouracil, azacitidine, cytarabine and irinotecan using a low concentration of sodium hypochlorite solution (0.115%). The analytical platform used to monitor the degradation course of the substances under examination was hydrogen nuclear magnetic spectroscopy (1H NMR). In the same experimental conditions the effectiveness of the degradation of the same antineoplastic drugs with a 99.9% ethanol solution was also evaluated. The study showed that the best degradation efficiency (> 90% ) is obtained with the hypochlorite solution after 15 minutes.


Subject(s)
Antineoplastic Agents/chemistry , Carcinogens/chemistry , Magnetic Resonance Spectroscopy/methods , Sodium Hypochlorite/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/classification , Azacitidine/chemistry , Carcinogens/classification , Cytarabine/chemistry , Decontamination/methods , Drug Interactions , Ethanol/pharmacology , Fluorouracil/chemistry , Humans , Hypochlorous Acid/pharmacology , Irinotecan/chemistry , Occupational Exposure/adverse effects , Occupational Exposure/prevention & control , Preliminary Data , Sodium Chloride/pharmacology , Time Factors
5.
Cell Biochem Funct ; 35(8): 488-496, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29143344

ABSTRACT

Cervical cancer is one of the most common malignancies of the female reproductive system. Therefore, it is critical to investigate the molecular mechanisms involved in the development and progression of cervical cancer. In this study, we stimulated cervical cancer cells with 5-aza-2'-deoxycytidine (5-Aza-dC) and found that this treatment inhibited cell proliferation and induced apoptosis; additionally, methylation of p16 and O-6-methylguanine-DNA methyltransferase (MGMT) was reversed, although their expression was suppressed. 5-Aza-dC inhibited E6 and E7 expression and up-regulated p53, p21, and Rb expression. Cells transfected with siRNAs targeting p16 and MGMT as well as cells stimulated with 5-Aza-dC were arrested in S phase, and the expression of p53, p21, and Rb was up-regulated more significantly. However, when cells were stimulated with 5-Aza-dC after transfection with siRNAs targeting p16 and MGMT, proliferation decreased significantly, and the percentage of cells in the sub-G1 peak and in S phase was significantly increased, suggesting a marked increase in apoptosis. But E6 and E7 overexpression could rescue the observed effects in proliferation. Furthermore, X-ray radiation caused cells to arrest in G2/M phase, but cells transfected with p16- and MGMT-targeted siRNAs followed by X-ray radiation exhibited a significant decrease in proliferation and were shifted toward the sub-G1 peak, also indicating enhanced apoptosis. In addition, the effects of 5-Aza-dC and X-ray radiation were most pronounced when MGMT expression was down-regulated. Therefore, down-regulation of p16 and MGMT expression enhances the anti-proliferative effects of 5-Aza-dC and X-ray radiation. This discovery may provide novel ideas for the treatment of cervical cancer.


Subject(s)
Apoptosis/drug effects , Azacitidine/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p18/antagonists & inhibitors , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Down-Regulation/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Azacitidine/chemistry , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Decitabine , Down-Regulation/genetics , Female , Humans , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , X-Rays
6.
Proc Natl Acad Sci U S A ; 111(32): E3252-9, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071207

ABSTRACT

Viral lethal mutagenesis is a strategy whereby the innate immune system or mutagenic pool nucleotides increase the error rate of viral replication above the error catastrophe limit. Lethal mutagenesis has been proposed as a mechanism for several antiviral compounds, including the drug candidate 5-aza-5,6-dihydro-2'-deoxycytidine (KP1212), which causes A-to-G and G-to-A mutations in the HIV genome, both in tissue culture and in HIV positive patients undergoing KP1212 monotherapy. This work explored the molecular mechanism(s) underlying the mutagenicity of KP1212, and specifically whether tautomerism, a previously proposed hypothesis, could explain the biological consequences of this nucleoside analog. Establishing tautomerism of nucleic acid bases under physiological conditions has been challenging because of the lack of sensitive methods. This study investigated tautomerism using an array of spectroscopic, theoretical, and chemical biology approaches. Variable temperature NMR and 2D infrared spectroscopic methods demonstrated that KP1212 existed as a broad ensemble of interconverting tautomers, among which enolic forms dominated. The mutagenic properties of KP1212 were determined empirically by in vitro and in vivo replication of a single-stranded vector containing a single KP1212. It was found that KP1212 paired with both A (10%) and G (90%), which is in accord with clinical observations. Moreover, this mutation frequency is sufficient for pushing a viral population over its error catastrophe limit, as observed before in cell culture studies. Finally, a model is proposed that correlates the mutagenicity of KP1212 with its tautomeric distribution in solution.


Subject(s)
Anti-HIV Agents/pharmacology , Azacitidine/analogs & derivatives , Deoxycytidine/analogs & derivatives , HIV/drug effects , HIV/genetics , Mutagens/pharmacology , Anti-HIV Agents/chemistry , Azacitidine/chemistry , Azacitidine/pharmacology , Bacteriophage M13/drug effects , Bacteriophage M13/genetics , Bacteriophage M13/physiology , Base Pairing , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Genome, Viral/drug effects , HIV/physiology , Humans , Isomerism , Magnetic Resonance Spectroscopy , Models, Chemical , Mutagens/chemistry , Spectrophotometry, Infrared , Virus Replication/drug effects , Virus Replication/genetics
7.
J Bacteriol ; 198(20): 2776-83, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27457718

ABSTRACT

UNLABELLED: We tested pairwise combinations of classical base analog mutagens in Escherichia coli to study possible mutagen synergies. We examined the cytidine analogs zebularine (ZEB) and 5-azacytidine (5AZ), the adenine analog 2-aminopurine (2AP), and the uridine/thymidine analog 5-bromodeoxyuridine (5BrdU). We detected a striking synergy with the 2AP plus ZEB combination, resulting in hypermutability, a 35-fold increase in mutation frequency (to 53,000 × 10(-8)) in the rpoB gene over that with either mutagen alone. A weak synergy was also detected with 2AP plus 5AZ and with 5BrdU plus ZEB. The pairing of 2AP and 5BrdU resulted in suppression, lowering the mutation frequency of 5BrdU alone by 6.5-fold. Sequencing the mutations from the 2AP plus ZEB combination showed the predominance of two new hot spots for A·T→G·C transitions that are not well represented in either single mutagen spectrum, and one of which is not found even in the spectrum of a mismatch repair-deficient strain. The strong synergy between 2AP and ZEB could be explained by changes in the dinucleoside triphosphate (dNTP) pools. IMPORTANCE: Although mutagens have been widely studied, the mutagenic effects of combinations of mutagens have not been fully researched. Here, we show that certain pairwise combinations of base analog mutagens display synergy or suppression. In particular, the combination of 2-aminopurine and zebularine, analogs of adenine and cytidine, respectively, shows a 35-fold increased mutation frequency compared with that of either mutagen alone. Understanding the mechanism of synergy can lead to increased understanding of mutagenic processes. As combinations of base analogs are used in certain chemotherapy regimens, including those involving ZEB and 5AZ, these results indicate that testing the mutagenicity of all drug combinations is prudent.


Subject(s)
Azacitidine/toxicity , Base Pairing/drug effects , Bromodeoxyuridine/toxicity , Cytidine/analogs & derivatives , Escherichia coli/drug effects , Mutagens/toxicity , Mutation/drug effects , Azacitidine/chemistry , Bromodeoxyuridine/chemistry , Cytidine/chemistry , Cytidine/toxicity , Drug Synergism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mutagens/chemistry
8.
J Biol Chem ; 290(3): 1829-36, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25492871

ABSTRACT

Regulation of transcription elongation by positive transcription elongation factor b (P-TEFb) plays a central role in determining the state of cell activation, proliferation, and differentiation. In cells, P-TEFb exists in active and inactive forms. Its release from the inactive 7SK small nuclear ribonucleoprotein complex is a critical step for P-TEFb to activate transcription elongation. However, no good method exists to analyze this P-TEFb equilibrium in living cells. Only inaccurate and labor-intensive cell-free biochemical assays are currently available. In this study, we present the first experimental system to monitor P-TEFb activation in living cells. We created a bimolecular fluorescence complementation assay to detect interactions between P-TEFb and its substrate, the C-terminal domain of RNA polymerase II. When cells were treated with suberoylanilide hydroxamic acid, which releases P-TEFb from the 7SK small nuclear ribonucleoprotein, they turned green. Other known P-TEFb-releasing agents, including histone deacetylase inhibitors, bromodomain and extraterminal bromodomain inhibitors, and protein kinase C agonists, also scored positive in this assay. Finally, we identified 5'-azacytidine as a new P-TEFb-releasing agent. This release of P-TEFb correlated directly with activation of human HIV and HEXIM1 transcription. Thus, our visualization of P-TEFb activation by fluorescent complementation assay could be used to find new P-TEFb-releasing agents, compare different classes of agents, and assess their efficacy singly and/or in combination.


Subject(s)
Positive Transcriptional Elongation Factor B/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Apoptosis , Azacitidine/chemistry , Bacterial Proteins/chemistry , Cell-Free System , Genetic Complementation Test , Glycerol/chemistry , HEK293 Cells , HeLa Cells , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemistry , Luminescent Proteins/chemistry , Microscopy, Fluorescence , Plasmids/metabolism , Protein Kinase C/metabolism , Protein Structure, Tertiary , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Time Factors , Transcription, Genetic , Vorinostat
9.
Blood ; 124(26): 3914-23, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25359989

ABSTRACT

Therapeutic natural killer (NK)-cell-mediated alloreactivity toward acute myeloid leukemia has largely been attributed to mismatches between killer immunoglobulin-like receptors (KIRs) on NK cells and their ligands, HLA class I molecules, on target cells. While adult acute B-cell precursor leukemia (BCP-ALL) appears to be resistant to NK-cell-mediated lysis, recent data indicate that pediatric BCP-ALL might yet be a target of NK cells. In this study, we demonstrate in a donor-patient-specific NOD.Cg-Prkdc(scid) IL2rg(tmWjl)/Sz (NSG) xenotransplantation model that NK cells mediate considerable alloreactivity toward pediatric BCP-ALL in vivo. Notably, both adoptively transferred mature KIR(+) NK cells and immature KIR(-) NK cells arising early posttransplantation in humanized NSG mice exerted substantial antileukemic activity. Low-dose and long-term treatment of humanized NSG mice with the DNA-demethylating agent 5-aza-cytidine distinctly enhanced the antitumor response, interestingly without inducing common inhibitory KIR expression but rather by promoting the differentiation of various NK-cell precursor subsets. Collectively, these data indicate that the future design of innovative therapy protocols should consider further exploitation of NK-cell-mediated immune responses for poor prognosis pediatric BCP-ALL patients.


Subject(s)
Antineoplastic Agents/chemistry , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/cytology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, KIR/metabolism , Animals , Azacitidine/chemistry , Child , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , DNA Methylation , Disease Models, Animal , Genotype , Graft vs Leukemia Effect , Humans , Interleukin-2/genetics , Mice , Mice, Inbred NOD , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , Transplantation, Heterologous
10.
Blood ; 123(4): 554-61, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24311721

ABSTRACT

CD33 is a valid target for acute myeloid leukemia (AML) but has proven challenging for antibody-drug conjugates. Herein, we investigated the cellular determinants for the activity of the novel CD33/CD3-directed bispecific T-cell engager antibody, AMG 330. In the presence of T cells, AMG 330 was highly active against human AML cell lines and primary AML cells in a dose- and effector to target cell ratio-dependent manner. Using cell lines engineered to express wild-type CD33 at increased levels, we found a quantitative relationship between AMG 330 cytotoxicity and CD33 expression; in contrast, AMG 330 cytotoxicity was neither affected by common CD33 single nucleotide polymorphisms nor expression of the adenosine triphosphate-binding cassette (ABC) transporter proteins, P-glycoprotein or breast cancer resistance protein. Unlike bivalent CD33 antibodies, AMG 330 did not reduce surface CD33 expression. The epigenetic modifier drugs, panobinostat and azacitidine, increased CD33 expression in some cell lines and augmented AMG 330-induced cytotoxicity. These findings demonstrate that AMG 330 has potent CD33-dependent cytolytic activity in vitro, which can be further enhanced with other clinically available therapeutics. As it neither modulates CD33 expression nor is affected by ABC transporter activity, AMG 330 is highly promising for clinical exploration as it may overcome some limitations of previous CD33-targeted agents.


Subject(s)
Antibodies, Bispecific/chemistry , Enzyme Inhibitors/chemistry , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , T-Lymphocytes/cytology , AC133 Antigen , Antibodies/chemistry , Antigens, CD/metabolism , Azacitidine/chemistry , CD3 Complex/metabolism , Cell Line, Tumor , Epigenesis, Genetic , Glycoproteins/metabolism , HL-60 Cells , Humans , Hydroxamic Acids/chemistry , Indoles/chemistry , Leukocytes, Mononuclear/cytology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Panobinostat , Peptides/metabolism , Polymorphism, Single Nucleotide , T-Lymphocytes/metabolism
11.
Bioorg Med Chem ; 24(11): 2410-22, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27117260

ABSTRACT

Although many compounds have been approved for the treatment of human immunodeficiency type-1 (HIV-1) infection, additional anti-HIV-1 drugs (particularly those belonging to new drug classes) are still needed due to issues such as long-term drug-associated toxicities, transmission of drug-resistant variants, and development of multi-class resistance. Lethal mutagenesis represents an antiviral strategy that has not yet been clinically translated for HIV-1 and is based on the use of small molecules to induce excessive levels of deleterious mutations within the viral genome. Here, we show that 5-azacytidine (5-aza-C), a ribonucleoside analog that induces the lethal mutagenesis of HIV-1, and multiple inhibitors of the enzyme ribonucleotide reductase (RNR) interact in a synergistic fashion to more effectively reduce the infectivity of HIV-1. In these drug combinations, RNR inhibitors failed to significantly inhibit the conversion of 5-aza-C to 5-aza-2'-deoxycytidine, suggesting that 5-aza-C acts primarily as a deoxyribonucleoside even in the presence of RNR inhibitors. The mechanism of antiviral synergy was further investigated for the combination of 5-aza-C and one specific RNR inhibitor, resveratrol, as this combination improved the selectivity index of 5-aza-C to the greatest extent. Antiviral synergy was found to be primarily due to the reduced accumulation of reverse transcription products rather than the enhancement of viral mutagenesis. To our knowledge, these observations represent the first demonstration of antiretroviral synergy between a ribonucleoside analog and RNR inhibitors, and encourage the development of additional ribonucleoside analogs and RNR inhibitors with improved antiretroviral activity.


Subject(s)
Anti-HIV Agents/pharmacology , Azacitidine/pharmacology , Enzyme Inhibitors/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Ribonucleotide Reductases/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Azacitidine/chemical synthesis , Azacitidine/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Microbial Sensitivity Tests , Molecular Structure , Ribonucleotide Reductases/metabolism , Structure-Activity Relationship
12.
J Reprod Dev ; 62(1): 71-7, 2016.
Article in English | MEDLINE | ID: mdl-26537205

ABSTRACT

Nuclear reprogramming induced by somatic cell nuclear transfer is an inefficient process, and donor cell DNA methylation status is thought to be a major factor affecting cloning efficiency. Here, the role of donor cell DNA methylation status regulated by 5-aza-2'-deoxycytidine (5-aza-dC) or 5-methyl-2'-deoxycytidine-5'-triphosphate (5-methyl-dCTP) in the early development of porcine cloned embryos was investigated. Our results showed that 5-aza-dC or 5-methyl-dCTP significantly reduced or increased the global methylation levels and altered the methylation and expression levels of key genes in donor cells. However, the development of cloned embryos derived from these cells was reduced. Furthermore, disrupted pseudo-pronucleus formation and transcripts of early embryo development-related genes were observed in cloned embryos derived from these cells. In conclusion, our results demonstrated that alteration of the DNA methylation status of donor cells by 5-aza-dC or 5-methyl-dCTP disrupted nuclear reprogramming and impaired the developmental competence of porcine cloned embryos.


Subject(s)
Cloning, Organism/methods , DNA Methylation , Animals , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Blastocyst/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cellular Reprogramming , Decitabine , Deoxycytosine Nucleotides/chemistry , Embryo Culture Techniques , Embryonic Development/drug effects , Fertilization in Vitro , Fibroblasts/metabolism , Genome , Oocytes/cytology , Swine
13.
Biomed Chromatogr ; 30(3): 494-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26174363

ABSTRACT

The DNA methyltransferase inhibitor 5-azacytidine is being evaluated clinically as an oral formulation to treat various solid tumors. A sensitive, reliable method was developed to quantitate 5-azacytidine using LC-MS/MS to perform detailed pharmacokinetic studies. The drug of interest was extracted from plasma using Oasis MCX ion exchange solid-phase extraction 96-well plates. Chromatographic separation was achieved with a YMC J'sphere M80 C18 column and isocratic elution with a methanol-water-formic acid (15:85:0.1, v/v/v) mobile phase over a 7 min total analytical run time. An AB Sciex 5500 triple quadrupole mass spectrometer operated in positive electrospray ionization mode was used for the detection of 5-azacytidine. The assay range was 5-500 ng/mL and proved to be accurate (97.8-109.1%) and precise (CV ≤ 9.8%). Tetrahydrouridine was used to stabilize 5-azacytidine in blood/plasma samples. With the addition of tetrahydrouridine, long-term frozen plasma stability for 5-azacytidine at -70°C has been determined for at least 323 days. The method was applied for the measurement of total plasma concentrations of 5-azacytidine in a cancer patient receiving a 300 mg oral daily dose.


Subject(s)
Antimetabolites, Antineoplastic/blood , Azacitidine/blood , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/chemistry , Azacitidine/pharmacokinetics , Azacitidine/therapeutic use , Drug Stability , Humans , Neoplasms/drug therapy , Reproducibility of Results , Sensitivity and Specificity
14.
J Biol Chem ; 289(46): 32276-32290, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25266720

ABSTRACT

Estrogen-related receptor α (ESRRA) functions as a transcription factor and regulates the expression of several genes, such as WNT11 and OPN. Up-regulation of ESRRA has been reported in several cancers. However, the mechanism underlying its up-regulation is unclear. Furthermore, the reports regarding the role and regulation of ESRRA in oral squamous cell carcinoma (OSCC) are completely lacking. Here, we show that tumor suppressor miR-125a directly binds to the 3'UTR of ESRRA and represses its expression. Overexpression of miR-125a in OSCC cells drastically reduced the level of ESRRA, decreased cell proliferation, and increased apoptosis. Conversely, the delivery of an miR-125a inhibitor to these cells drastically increased the level of ESRRA, increased cell proliferation, and decreased apoptosis. miR-125a-mediated down-regulation of ESRRA impaired anchorage-independent colony formation and invasion of OSCC cells. Reduced cell proliferation and increased apoptosis of OSCC cells were dependent on the presence of the 3'UTR in ESRRA. The delivery of an miR-125a mimic to OSCC cells resulted in marked regression of xenografts in nude mice, whereas the delivery of an miR-125a inhibitor to OSCC cells resulted in a significant increase of xenografts and abrogated the tumor suppressor function of miR-125a. We observed an inverse correlation between the expression levels of miR-125a and ESRRA in OSCC samples. In summary, up-regulation of ESRRA due to down-regulation of miR-125a is not only a novel mechanism for its up-regulation in OSCC, but decreasing the level of ESRRA by using a synthetic miR-125a mimic may have an important role in therapeutic intervention of OSCC and other cancers.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Mouth Neoplasms/metabolism , Amino Acid Sequence , Animals , Apoptosis , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Cell Line, Tumor , Cell Proliferation , Decitabine , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Transplantation , Plasmids/metabolism , Protein Binding , Sequence Homology, Amino Acid
15.
Biochem Biophys Res Commun ; 456(1): 344-50, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25475733

ABSTRACT

Phosphatase of regenerating liver-3 (PRL-3) is believed to be associated with cell motility, invasion, and metastasis. Our previous work found that PRL-3 is highly overexpressed in gastric cancer (GC) tissue with peritoneal metastasis and directly involved in the pathogenesis of GC peritoneal metastasis. Moreover, we further found that the down-regulation of endogenous miR-495 expression plays a causative role in over expression of PRL-3 in GC peritoneal metastasis. However, the molecular regulation mechanisms by which endogenous miR-495 expression is down-regulated and PRL-3 promotes GC peritoneal metastasis remain to be clearly elucidated. Some studies have shown that the promoter methylation is closely related to the miRNA gene expression. Therefore, in present study, based on our previous findings, we will analysis whether DNA methylation is a major cause of the down-expression of endogenous miR-495, which results in PRL-3 overexpression in GC peritoneal metastasis. Methylation specific PCR (MSP) and sodium bisulfite sequencing method (BSP) detected miR-495 gene promoter methylation status. We treated GC cell lines with 5-Aza-2'-deoxycytidine (5-Aza-dC) to make the gene promoter methylation inactivation. By treating with 5-Aza-dC the migration and invasion of GC cells were significantly inhibited. And the miR-495 was overexpressing, corresponds to the mRNA and protein levels of PRL-3 were reduced, the ability of invasion and metastasis was inhibited. This study suggest that miR-495 have tumor suppressor properties and are partially silenced by DNA hypermethylation in GC, will provide new strategies for prevention and treatment of GC peritoneal metastasis.


Subject(s)
DNA Methylation , Gene Silencing , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Protein Tyrosine Phosphatases/metabolism , Stomach Neoplasms/metabolism , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Cell Line, Tumor , Cell Movement , Decitabine , Genes, Tumor Suppressor , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Sequence Analysis, DNA
16.
Mol Pharm ; 12(4): 1279-88, 2015 Apr 06.
Article in English | MEDLINE | ID: mdl-25751281

ABSTRACT

In this study, we developed and characterized a delivery system for the epigenetic demethylating drug, decitabine, to sensitize temozolomide-resistant human glioblastoma multiforme (GBM) cells to alkylating chemotherapy. A poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) based nanoconjugate was fabricated to encapsulate decitabine and achieved a better therapeutic response in GBM cells than that with the free drug. After synthesis, the highly efficient uptake process and intracellular dynamics of this nanoconjugate were monitored by single-molecule fluorescence tools. Our experiments demonstrated that, under an acidic pH due to active glycolysis in cancer cells, the PLGA-PEG nanovector could release the conjugated decitabine at a faster rate, after which the hydrolyzed lactic acid and glycolic acid would further acidify the intracellular microenvironment, thus providing positive feedback to increase the effective drug concentration and realize growth inhibition. In temozolomide-resistant GBM cells, decitabine can potentiate the cytotoxic DNA alkylation by counteracting cytosine methylation and reactivating tumor suppressor genes, such as p53 and p21. Owing to the excellent internalization and endolysosomal escape enabled by the PLGA-PEG backbone, the encapsulated decitabine exhibited a better anti-GBM potential than that of free drug molecules. Hence, the synthesized nanoconjugate and temozolomide could act in synergy to deliver a more potent and long-term antiproliferative effect against malignant GBM cells.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Azacitidine/analogs & derivatives , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Nanoconjugates/chemistry , Antimetabolites, Antineoplastic/chemistry , Apoptosis , Azacitidine/chemistry , Biocompatible Materials/chemistry , Boron Compounds , Cell Line, Tumor , DNA Methylation , Dacarbazine/administration & dosage , Decitabine , Drug Delivery Systems , Drug Synergism , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Lactic Acid/chemistry , Magnetic Resonance Spectroscopy , Methacrylates , Methylmethacrylates , Micelles , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Spectrometry, Fluorescence , Temozolomide
17.
Biochem J ; 463(3): 329-37, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25101795

ABSTRACT

During the course of protein synthesis in the cell, the translation process is often terminated due to various reasons. As a result, peptidyl-tRNA molecules are released which are toxic to the cell as well reducing the availability of free amino acid and tRNA molecules for the required protein synthesis in the cell. Such a situation is corrected by an enzyme, Pth (peptidyl-tRNA hydrolase), which catalyses the release of free tRNA and peptide moieties from peptidyl-tRNAs. This means that the active Pth is essential for the survival of bacteria. In order to design inhibitors of PaPth (Pth from Pseudomonas aeruginosa), we determined the structures of PaPth in its native and bound states with compounds amino acylate-tRNA analogue and 5-azacytidine. The structure determination of the native protein revealed that the substrate-binding site was partially occupied by Glu161 from the neigh-bouring molecule. The structure of PaPth indicated that the substrate-binding site can be broadly divided into three distinct subsites. The structures of the two complexes showed that the amino acylate-tRNA analogue filled three subsites, whereas 5-azacytidine filled two subsites. The common sugar and the base moieties of the two compounds occupied identical positions in the cleft. Using surface plasmon resonance, the dissociation constants for the amino acylate-tRNA analogue and 5-azacytidine were found to be 3.53×10-8 M and 5.82×10-8 M respectively.


Subject(s)
Adenosine/analogs & derivatives , Bacterial Proteins/chemistry , Carboxylic Ester Hydrolases/chemistry , Enzyme Inhibitors/chemistry , Pseudomonas aeruginosa/enzymology , Tyrosine/analogs & derivatives , Adenosine/chemistry , Azacitidine/chemistry , Carboxylic Ester Hydrolases/antagonists & inhibitors , Catalytic Domain , Crystallography, X-Ray , Drug Design , Hydrogen Bonding , Protein Conformation , Structure-Activity Relationship , Tyrosine/chemistry
18.
Nucleic Acids Res ; 41(9): e104, 2013 May.
Article in English | MEDLINE | ID: mdl-23519618

ABSTRACT

A number of proteins form covalent bonds with DNA as obligatory transient intermediates in normal nuclear transactions. Drugs that trap these complexes have proven to be potent therapeutics in both cancer and infectious disease. Nonetheless, current assays for DNA-protein adducts are cumbersome, limiting both mechanistic studies and translational applications. We have developed a rapid and sensitive assay that enables quantitative immunodetection of protein-DNA adducts. This new 'RADAR' (rapid approach to DNA adduct recovery) assay accelerates processing time 4-fold, increases sample throughput 20-fold and requires 50-fold less starting material than the current standard. It can be used to detect topoisomerase 1-DNA adducts in as little as 60 ng of DNA, corresponding to 10 000 human cells. We apply the RADAR assay to demonstrate that expression of SLFN11 does not increase camptothecin sensitivity by promoting accumulation of topoisomerase 1-DNA adducts. The RADAR assay will be useful for analysis of the mechanisms of formation and resolution of DNA-protein adducts in living cells, and identification and characterization of reactions in which covalent DNA adducts are transient intermediates. The assay also has potential application to drug discovery and individualized medicine.


Subject(s)
DNA Adducts/analysis , DNA-Binding Proteins/analysis , Immunoassay/methods , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Camptothecin/toxicity , Cell Line , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/analysis , DNA (Cytosine-5-)-Methyltransferases/immunology , DNA Adducts/chemistry , DNA Topoisomerases, Type I/analysis , DNA Topoisomerases, Type I/immunology , DNA-Binding Proteins/chemistry , Decitabine , Humans , Nuclear Proteins/antagonists & inhibitors , Topoisomerase I Inhibitors/toxicity
19.
J Biol Chem ; 288(33): 23858-67, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23839987

ABSTRACT

Methylation of cytosines in CpG dinucleotides is the predominant epigenetic mark on vertebrate DNA. DNA methylation is associated with transcriptional repression. The pattern of DNA methylation changes during development and with disease. Human DNA methyltransferase 1 (Dnmt1), a 1616-amino acid multidomain enzyme, is essential for maintenance of DNA methylation in proliferating cells and is considered an important cancer drug target. Using a fluorogenic, endonuclease-coupled DNA methylation assay with an activated form of Dnmt1 engineered to lack the replication foci targeting sequence domain, we discovered that laccaic acid A (LCA), a highly substituted anthraquinone natural product, is a direct inhibitor with a 310 nm Ki. LCA is competitive with the DNA substrate in in vitro methylation assays and alters the expression of methylated genes in MCF-7 breast cancer cells synergistically with 5-aza-2'-deoxycytidine. LCA represents a novel class of Dnmt-targeted molecular probes, with biochemical properties that allow it to distinguish between non DNA-bound and DNA-bound Dnmt1.


Subject(s)
Azo Compounds/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA/metabolism , Enzyme Inhibitors/pharmacology , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Anthraquinones/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Azacitidine/pharmacology , Azo Compounds/chemistry , Base Sequence , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , DNA Replication/drug effects , Decitabine , Enzyme Inhibitors/chemistry , Female , Fluorometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Reproducibility of Results , Signal Transduction/drug effects , Signal Transduction/genetics , Transition Temperature
20.
Calcif Tissue Int ; 94(4): 414-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24322886

ABSTRACT

Mechanical stimulation reduces sclerostin expression in rodents. However, few data are available about the effect of physical stimuli in human systems. Recently we observed that the demethylating agent AzadC induces SOST expression in bone cells. This allowed us in this study to explore the effect of mechanical loading on SOST expression by subjecting AzadC-treated human bone cells to pulsating fluid flow (PFF). PFF significantly decreased the AzadC-induced expression of SOST. This effect persisted for at least 24 h, and in fact SOST expression was lower at 24 h after PFF treatment than at 1 h after PFF treatment (PFF/static ratio 0.47 ± 0.04 vs. 0.63 ± 0.03 respectively, p = 0.03). The PFF-induced decrease in SOST expression was not due to a change in the methylation profile of the SOST promoter. However, PFF stimulated nitric oxide (NO) synthesis, which appeared essential for the PFF effect on SOST expression. In fact, the NO synthase inhibitor 1400 W prevented the effect of PFF on SOST expression. Moreover, the NO-donor SNAP decreased SOST mRNA in bone organ cultures. The conditioned medium (CM) of cells subjected to PFF induced a 38 ± 4 % decrease in SOST expression (p = 0.03) in static cultures and diminished the transcriptional activity of reporter vectors with the cloned SOST promoter (Static-CM: 1.47 ± 0.10 vs. PFF-CM: 0.78 ± 0.09, p = 0.02). This is consistent with a PFF-induced secretion of factors that modulate SOST. Our results suggest that NO and other soluble factors are involved in the inhibition of SOST expression by PFF.


Subject(s)
Azacitidine/analogs & derivatives , Bone Morphogenetic Proteins/metabolism , Down-Regulation , Nitric Oxide/chemistry , Stress, Mechanical , Adaptor Proteins, Signal Transducing , Azacitidine/chemistry , Bone and Bones/metabolism , Cell Line , Culture Media, Conditioned/chemistry , DNA Methylation , Decitabine , Genetic Markers , Humans , Osteoblasts/drug effects , Pulsatile Flow , Signal Transduction , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL