Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Curr Mol Pharmacol ; 14(1): 60-67, 2021.
Article in English | MEDLINE | ID: mdl-32394848

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most fatal malignancies worldwide and despite using various therapeutic strategies for the treatment of HNSCC, the surveillance rate is low. Telomerase has been remarked as the primary target in cancer therapy. Considering the key regulatory role of epigenetic mechanisms in controlling genome expression, the present study aimed to investigate the effects of two epigenetic modulators, a DNA methylation inhibitor and a histone deacetylase inhibitor on cell migration, proliferation, hTERT gene expression, and telomerase activity in HNSCC cell lines. METHODS: Human HNSCC cell lines were treated with Azacitidine and Trichostatin A to investigate their effects on telomerase gene expression and activity. Cell viability, migration, hTERT gene expression, and telomerase activity were studied using MTT colorimetric assay, scratch wound assay, qRT-PCR, and TRAP assay, respectively. RESULTS: Azacitidine at concentrations of ≤1µM and Trichostatin A at 0.1 to 0.3nM concentrations significantly decreased FaDu and Cal-27 cells migration. The results showed that Azacitidine significantly decreased hTERT gene expression and telomerase activity in FaDu and Cal-27 cell lines. However, there were no significant changes in hTERT gene expression at different concentrations of Trichostatin A in both cell lines. Trichostatin A treatment affected telomerase activity at the high dose of 0.3 nM Trichostatin A. CONCLUSION: The findings revealed that unlike histone deacetylase inhibitor, Azacitidine as an inhibitor of DNA methylation decreases telomerase expression in HNSCC cells. This might suggest the potential role of DNA methyltransferase inhibitors in telomerase-based therapeutic approaches in squamous cell carcinoma.


Subject(s)
Antineoplastic Agents/chemistry , Azacitidine/chemistry , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Head and Neck Neoplasms/drug therapy , Histone Deacetylase Inhibitors/chemistry , Squamous Cell Carcinoma of Head and Neck/drug therapy , Telomerase/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Azacitidine/pharmacology , Cell Line, Tumor , Cell Survival , DNA Methylation , Drug Development , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Peptide Fragments/genetics , Signal Transduction
2.
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
3.
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
4.
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
5.
Anticancer Agents Med Chem ; 20(7): 887-896, 2020.
Article in English | MEDLINE | ID: mdl-32067621

ABSTRACT

BACKGROUND: Currently, most of the drugs used in clinical applications show their pharmacological influences by inhibiting or activating enzymes. Therefore, enzyme inhibitors have an essential place in the drug design for many diseases. OBJECTIVE: The current study aimed to contribute to this growing drug design field (i.e., medicine discovery and development) by analyzing enzyme-drug interactions. METHODS: For this reason, Paraoxonase-I (PON1) enzyme was purified from fresh human serum by using rapid chromatographic techniques. Additionally, the inhibition effects of some antineoplastic agents were researched on the PON1. RESULTS: The enzyme was obtained with a specific activity of 2603.57 EU/mg protein. IC50 values for pemetrexed disodium, irinotecan hydrochloride, dacarbazine, and azacitidine were determined to be 9.63µM, 30.13µM, 53.31µM, and 21.00mM, respectively. These agents found to strongly inhibit PON1, with Ki constants ranging from 8.29±1.47µM to 23.34±2.71mM. Dacarbazine and azacitidine showed non-competitive inhibition, while other drugs showed competitive inhibition. Furthermore, molecular docking was performed using maestro for these agents. Among these, irinotecan hydrochloride and pemetrexed disodium possess the binding energy of -5.46 and -8.43 kcal/mol, respectively. CONCLUSION: The interaction studies indicated that these agents with the PON1 possess binding affinity.


Subject(s)
Antineoplastic Agents/pharmacology , Aryldialkylphosphatase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Antineoplastic Agents/chemistry , Aryldialkylphosphatase/blood , Aryldialkylphosphatase/metabolism , Azacitidine/chemistry , Azacitidine/pharmacology , Dacarbazine/chemistry , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Design , Enzyme Inhibitors/chemistry , Humans , Irinotecan/chemistry , Irinotecan/pharmacology , Molecular Structure
6.
J Biomater Sci Polym Ed ; 31(1): 123-140, 2020 01.
Article in English | MEDLINE | ID: mdl-31594474

ABSTRACT

India has an alarming rate of growth of cardiovascular diseases (CVD). Similar to cancer there is a significant role for epigenetic factors in the increasing prevalence of CVD. Targeting the epigenetic mechanism, viz., the DNA methylation processes, histone modifications, and RNA based arrangements is today considered as a potential therapeutic approach to CVD management. 5-Azacytidine is an epigenetic treatment drug that is involved in the demethylation of DNA. 5-Azacytidine is an FDA approved drug for myelodysplastic syndrome. However, the usage of 5-Azacytidine for CVD has not been found acceptable because of its poor stability in neutral solutions and shorter half-live which makes it toxic to the cells. A significant breakthrough in the use of 5-azacytidine for cell therapy and tissue engineering for CVD treatment has been gained based on its ability to differentiate mesenchymal stem cells into cardiomyocytes. This work addresses the further need for a sustained release of this drug, to reduce its toxicity to the stem cells. Electrospun PCL-gelatin fibres that are well aligned to provide a mat-like structure with sufficient porosity for differentiated cells to move forward have been synthesized. The crystalline character, porosity, fibre width, thermal behavior hydrophilicity of these scaffolds are in tune with those reported in the literature as ideal for cell proliferation and adhesion. FTIR measurements confirm the entrapment of 5-azacytidine on to the scaffold. The adsorption of the drug did not alter the characteristic features of the scaffold. Primary results on cell viability and cell morphology, as well as cardiomyocyte differentiation, have shown that PCL-gelatin scaffolds carrying 5-azacytidine developed in this work could serve as an ideal platform for mesenchymal stem cell differentiation into cardiomyocytes.


Subject(s)
Azacitidine/chemistry , Azacitidine/pharmacology , Cell Differentiation/drug effects , Gelatin/chemistry , Myocytes, Cardiac/cytology , Nanostructures/chemistry , Polyesters/chemistry , Cell Line , Cell Proliferation/drug effects , Humans , Myocytes, Cardiac/drug effects
7.
Clin Epigenetics ; 11(1): 111, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31370878

ABSTRACT

BACKGROUND: DNA demethylation therapy is now used in practice for hematological tumors and is being developed for solid tumors. Nevertheless, it is difficult to achieve stable pharmacokinetics with the current DNA-demethylating agents, azacitidine (AZA) and decitabine (DAC), because of their rapid deamination by cytidine deaminase in vivo and spontaneous hydrolytic cleavage. Here, we aimed to develop metabolically stable prodrugs of AZA and DAC as novel DNA-demethylating agents. RESULTS: Thirty-five 5'-O-trialkylsilylated AZAs/DACs were synthesized with potential resistance to deamination. Out of these, 11 compounds exhibited demethylating activity similar to that of DAC and guadecitabine, and a suitable aqueous solubility. Pharmacokinetic analysis in mice showed that OR-2003 displayed the highest serum concentration and the area under the curve in an intraperitoneal experiment, whereas OR-2100 exhibited high stability to cytidine deaminase. Treatment of cells with OR-2003 and OR-2100 depleted DNA methyltransferase 1 completely and induced both gene-specific and genome-wide demethylation. The treatment suppressed the growth of multiple types of cancer cells and induced re-expression of tumor suppressor genes. The anti-tumor effect and DNA demethylation effect of OR-2003 and OR-2100 were comparable to that of DAC with fewer adverse effects in vivo. CONCLUSIONS: We developed two novel prodrugs of DAC that exhibited greater stability, comparable DNA demethylation activity, and less toxicity. These compounds are expected to overcome the difficulty in achieving stable pharmacokinetics in patients, leading to maximum DNA demethylation activity with minimum adverse effects.


Subject(s)
DNA Methylation/drug effects , Decitabine/chemistry , Neoplasms/drug therapy , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Animals , Area Under Curve , Azacitidine/chemistry , Blood Chemical Analysis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Stability , Gene Expression Regulation, Neoplastic/drug effects , Injections, Intraperitoneal , Mice , Neoplasms/genetics , Prodrugs/administration & dosage , Prodrugs/chemistry , Xenograft Model Antitumor Assays
8.
Article in English | MEDLINE | ID: mdl-30754019

ABSTRACT

Guadecitabine (SGI-110), a dinucleotide of ߭decitabine and deoxyguanosine, is currently being evaluated in phase II/III clinical trials for the treatment of hematological malignancies and solid tumors. This article describes the development and validation of bioanalytical assays to quantify guadecitabine and its active metabolite ߭decitabine in human plasma, whole blood and urine using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Since ߭decitabine is rapidly metabolized further by cytidine deaminase, plasma and whole blood samples were kept on ice-water after collection and stabilized with tetrahydrouridine (THU) directly upon sample collection. Sample preparation consisted of protein precipitation for plasma and whole blood and dilution for urine samples and was further optimized for each matrix and analyte separately. Final extracts were injected onto a C6-phenyl column for guadecitabine analysis, or a Nova-Pak Silica column for ߭decitabine analysis. Gradient elution was applied for both analytes using the same eluents for each assay and detection was performed on triple quadrupole mass spectrometers operating in the positive ion mode (Sciex QTRAP 5500 and QTRAP 6500). The assay for guadecitabine was linear over a range of 1.0-200 ng/mL (plasma, whole blood) and 10-2000 ng/mL (urine). For ߭decitabine the assay was linear over a range of 0.5-100 ng/mL (plasma, whole blood) and 5-1000 ng/mL (urine). The presented methods were successfully validated according to the latest FDA and EMA guidelines for bioanalytical method validation and applied in a guadecitabine clinical mass balance trial in patients with advanced cancer.


Subject(s)
Antineoplastic Agents/blood , Azacitidine/analogs & derivatives , Chromatography, Liquid/methods , Decitabine/blood , Tandem Mass Spectrometry/methods , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/urine , Azacitidine/blood , Azacitidine/chemistry , Azacitidine/pharmacokinetics , Azacitidine/urine , Decitabine/chemistry , Decitabine/pharmacokinetics , Decitabine/urine , Humans , Linear Models , Reproducibility of Results , Sensitivity and Specificity
9.
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
10.
J Pharm Biomed Anal ; 164: 16-26, 2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30366147

ABSTRACT

DNA hypermethylation is an epigenetic event that is commonly found in malignant cells and is used as a therapeutic target for ß-decitabine (ß-DEC) containing hypomethylating agents (eg Dacogen® and guadecitabine). ß-DEC requires cellular uptake and intracellular metabolic activation to ß-DEC triphosphate before it can get incorporated into the DNA. Once incorporated in the DNA, ß-DEC can exert its hypomethylating effect by trapping DNA methyltransferases (DNMTs), resulting in reduced 5-methyl-2'-deoxycytidine (5mdC) DNA content. ß-DEC DNA incorporation and its effect on DNA methylation, however, have not yet been investigated in patients treated with ß-DEC containing therapies. For this reason, we developed and validated a sensitive and selective LC-MS/MS method to determine total intracellular ß-DEC nucleotide (ß-DEC-XP) concentrations, as well as to quantify ß-DEC and 5mdC DNA incorporation relative to 2'-deoxycytidine (2dC) DNA content. The assay was successfully validated according to FDA and EMA guidelines in a linear range from 0.5 to 100 ng/mL (ß-DEC), 50 to 10,000 ng/mL (2dC), and 5 to 1,000 ng/mL (5mdC) in peripheral blood mononuclear cell (PBMC) lysate. An additional calibrator at a concentration of 0.1 ng/mL was added for ß-DEC to serve as a limit of detection (LOD). Clinical applicability of the method was demonstrated in patients treated with guadecitabine. Our data support the use of the validated LC-MS/MS method to further explore the intracellular pharmacokinetics in patients treated with ß-DEC containing hypomethylating agents.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Azacitidine/analogs & derivatives , DNA/chemistry , Decitabine/analysis , Deoxycytidine/analogs & derivatives , Adult , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/chemistry , Azacitidine/pharmacokinetics , Azacitidine/therapeutic use , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Clinical Trials, Phase II as Topic , DNA/metabolism , DNA Methylation/drug effects , Decitabine/chemistry , Deoxycytidine/analysis , Deoxycytidine/chemistry , Humans , Leukocytes, Mononuclear , Limit of Detection , Neoplasms/blood , Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Tandem Mass Spectrometry/instrumentation , Tandem Mass Spectrometry/methods
11.
Chem Biol Interact ; 296: 1-8, 2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30125549

ABSTRACT

BACKGROUD/AIMS: Abnormal activation of the Wnt/ß-catenin signaling, which may be antagonized by the members of secreted frizzled-related proteins family (SFRPs), is implicated in tumor occurrence and development. However, the function of SFRP5 relating to Wnt/ß-catenin pathway in chondrosarcoma is not clear yet. This study was undertaken to investigate the potential role of SFRP5 promoter methylation in chondrosarcoma metastasis and invasion through activating canonical Wnt signaling pathway. METHODS AND RESULTS: The results demonstrated that SFRP5 promoter was hypermethylated and SFRP5 expression was significantly reduced in chondrosarcoma cell lines at the mRNA and protein levels. The canonical Wnt/ß-catenin signaling was observably activated with ß-catenin stabilization by dephosphorylation and translocation into the nuclear. 5-Aza-2'-deoxycytidine (5-Aza-dC), the DNA methyltransferase inhibitor, significantly inhibited the proliferation of chondrosarcoma cells by cell cycle arrest through repressing the methylation of SFRP5 and promoting its expression. Both 5-Aza-dC treatment and SFRP5 overexpression could significantly inhibited the metastasis and invasion of chondrosarcoma cells by inactivating Wnt/ß-catenin signaling pathway and promoting chondrosarcoma cells mesenchymal-epithelial transition (MET). 5-Aza-dC also inhibited the xenograft growth and lung metastasis of chondrosarcoma cells in vivo via suppressing SFRP5 promotor methylation, inactivating Wnt/ß-catenin pathway and inducing epithelial markers expression. CONCLUSION: All of our results revealed the epigenetic silencing of SFRP5 by promoter methylation plays pivotal roles in chondrosarcoma development and metastasis through SFRP5/Wnt/ß-catenin signaling axis. Modulation of their levels may serve as potential targets and diagnostic tools for novel therapeutic strategies of chondrosarcoma.


Subject(s)
Chondrosarcoma/genetics , Chondrosarcoma/pathology , Epigenesis, Genetic/genetics , Eye Proteins/genetics , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Wnt Signaling Pathway/genetics , Adaptor Proteins, Signal Transducing , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Azacitidine/pharmacology , Cells, Cultured , Chondrosarcoma/drug therapy , Chondrosarcoma/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epigenesis, Genetic/drug effects , Eye Proteins/antagonists & inhibitors , Eye Proteins/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Structure-Activity Relationship , Wnt Signaling Pathway/drug effects
12.
Int J Pharm ; 541(1-2): 64-71, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29471144

ABSTRACT

Decitabine (DAC), a potent DNA methyltransferase (DNMT) inhibitor, has a limited oral bioavailability. Its 5'-amino acid ester prodrugs could improve its oral delivery but the specific absorption mechanism is not yet fully understood. The aim of this present study was to investigate the in vivo absorption and activation mechanism of these prodrugs using in situ intestinal perfusion and pharmacokinetics studies in rats. Although PEPT1 transporter is pH dependent, there appeared to be no proton cotransport in the perfusion experiment with a preferable transport at pH 7.4 rather than pH 6.5. This suggested that the transport was mostly dependent on the dissociated state of the prodrugs and the proton gradient might play only a limited role. In pH 7.4 HEPES buffer, an increase in Peff was observed for L-val-DAC, D-val-DAC, L-phe-DAC and L-trp-DAC (2.89-fold, 1.2-fold, 2.73-fold, and 1.90-fold, respectively), compared with the parent drug. When co-perfusing the prodrug with Glysar, a known substrate of PEPT1, the permeabilities of the prodrugs were significantly inhibited compared with the control. To further investigate the absorption of the prodrugs, L-val-DAC was selected and found to be concentration-dependent and saturable, suggesting a carrier-mediated process (intrinsic Km: 7.80 ±â€¯2.61 mM) along with passive transport. Determination of drug in intestinal homogenate after perfusion further confirmed that the metabolic activation mainly involved an intestinal first-pass effect. In a pharmacokinetic evaluation, the oral bioavailability of L-val-DAC, L-phe-DAC and L-trp-DAC were nearly 1.74-fold, 1.69-fold and 1.49-fold greater than that of DAC. The differences in membrane permeability and oral bioavailability might be due to the different stability in the intestinal lumen and the distinct PEPT1 affinity which is mainly caused by the stereochemistry, hydrophobicity and steric hindrance of the side chains. In summary, the detailed investigation of the absorption mechanism by in vivo intestinal perfusion and pharmacokinetic studies showed that the prodrugs of DAC exhibited excellent permeability and oral bioavailability, which might be attributed to a hybrid (partly PEPT1-mediated and partly passive) transport mode and a rapid activation process in enterocytes.


Subject(s)
Azacitidine/analogs & derivatives , Enterocytes/enzymology , Enzyme Inhibitors/pharmacokinetics , Intestinal Absorption , Peptide Transporter 1/metabolism , Prodrugs/pharmacokinetics , Administration, Oral , Amino Acids/chemistry , Animals , Azacitidine/administration & dosage , Azacitidine/chemistry , Azacitidine/pharmacokinetics , Biological Availability , Cell Membrane Permeability , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Esters/chemistry , Intestinal Mucosa/metabolism , Intestines/cytology , Male , Models, Animal , Prodrugs/administration & dosage , Prodrugs/chemistry , Rats , Rats, Sprague-Dawley
13.
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
14.
J Control Release ; 268: 92-101, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29042320

ABSTRACT

Myelodysplastic syndromes (MDS) are a diverse group of bone marrow disorders and clonal hematopoietic stem cell disorders characterized by abnormal blood cells, or reduced peripheral blood cell count. Recent clinical studies on combination therapy of decitabine (DAC) and arsenic trioxide (ATO) have demonstrated synergy on MDS treatment, but the treatment can cause significant side effects to patients. In addition, both drugs have to be administered on a daily basis due to their short half-lives. In addressing key issues of reducing toxic side effects and improving pharmacokinetic profiles of the therapeutic agents, we have developed a new formulation by co-packaging DAC and ATO into alendronate-conjugated bone-targeting nanoparticles (BTNPs). Our pharmacokinetic studies revealed that intravenously administered BTNPs increased circulation time up to 3days. Biodistribution analysis showed that the BTNP facilitated DAC and ATO accumulation in the bone, which is 6.7 and 7.9 times more than untargeted NP. Finally, MDS mouse model treated with BTNPs showed better restoration of complete blood count to normal level, and significantly longer median survival as compared to free drugs or untargeted NPs treatment. Our results support bone-targeted co-delivery of DAC and ATO for effective treatment of MDS.


Subject(s)
Antineoplastic Agents/administration & dosage , Arsenicals/administration & dosage , Azacitidine/analogs & derivatives , Myelodysplastic Syndromes/drug therapy , Nanoparticles/administration & dosage , Oxides/administration & dosage , Alendronate/administration & dosage , Alendronate/chemistry , Alendronate/pharmacokinetics , Alendronate/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/chemistry , Arsenicals/pharmacokinetics , Arsenicals/therapeutic use , Azacitidine/administration & dosage , Azacitidine/chemistry , Azacitidine/pharmacokinetics , Azacitidine/therapeutic use , Bone Marrow Cells/drug effects , Bone and Bones/metabolism , Cell Survival/drug effects , Cells, Cultured , Decitabine , Mice, Transgenic , Myelodysplastic Syndromes/metabolism , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/therapeutic use , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacokinetics , Phosphatidylethanolamines/therapeutic use , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/therapeutic use , Tissue Distribution
15.
Epigenetics ; 12(10): 886-896, 2017.
Article in English | MEDLINE | ID: mdl-28758855

ABSTRACT

In acute myeloid leukemia (AML) DNA hypermethylation of gene promoters is frequently observed and often correlates with a block of differentiation. Treatment of AML patients with DNA methyltransferase inhibitors results in global hypomethylation of genes and, thereby, can lead to a reactivation of the differentiation capability. Unfortunately, after termination of treatment both hypermethylation and differentiation block return in most cases. Here, we apply, for the first time, a computational model of epigenetic regulation of transcription to: i) provide a mechanistic understanding of the DNA (de-) methylation process in AML and; ii) improve DNA demethylation treatment strategies. By in silico simulation, we analyze promoter hypermethylation scenarios referring to DNMT dysfunction, decreased H3K4me3 and increased H3K27me3 modification activity, and accelerated cell proliferation. We quantify differences between these scenarios with respect to gene repression and activation. Moreover, we compare the scenarios regarding their response to DNMT inhibitor treatment alone and in combination with inhibitors of H3K27me3 histone methyltransferases and of H3K4me3 histone demethylases. We find that the different hypermethylation scenarios respond specifically to therapy, suggesting that failure of remission originates in patient-specific deregulation. We observe that inappropriate demethylation therapy can result even in enforced deregulation. As an example, our results suggest that application of high DNMT inhibitor concentration can induce unwanted global gene activation if hypermethylation originates in increased H3K27me3 modification. Our results underline the importance of a personalized therapy requiring knowledge about the patient-specific mechanism of epigenetic deregulation.


Subject(s)
DNA Demethylation/drug effects , DNA Methylation/genetics , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/genetics , Azacitidine/chemistry , Azacitidine/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/genetics , Computational Biology , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/genetics , Enzyme Inhibitors/chemistry , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/chemistry , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Models, Molecular , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics
16.
Comput Biol Chem ; 70: 49-55, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28802167

ABSTRACT

5'-Aza-2'-deoxycytidine (5-Aza-dC) is a demethylating drug that causes genome-wide hypomethylation resulting in the expression of several tumor suppressor genes causing growth arrest of cancer cells. Cancer is well established as a multifactorial disease and requires multi-module therapeutics. Search for new drugs and their approval by FDA takes a long time. Keeping this in view, research on new functions of FDA-approved anticancer drugs is desired to expand the list of multi-module functioning drugs for cancer therapy. In this study, we conducted an analysis for new functions of 5-Aza-dC by applying bio-chemo-informatics approach. The potential of 5-Aza-dC bioactivity was analyzed by PASS online and Molinspiration. Target proteins were predicted by SuperPred. The protein networks and biological processes were analyzed by Biological Networks using Gene Ontology tool, BINGO, based on BIOGRID database. Interactions between 5-Aza-dC and targeted proteins were examined by Autodoc Vina integrated into pyrx software. Induction of p53 by 5-Aza-dC was tested in vitro using cancer cells. Bioinformatics analyses predicted that 5-Aza-dC functions as a p53 inducer, radiosensitizer, and inhibitor of some enzymes. It was predicted to target proteins including MDM2, POLA1, POLB, and CXCR4 that are involved in the induction of DNA damage response and p53-HDM2-p21 signaling. In this study, we provide experimental evidence showing HDM2 is one of the targets of 5-AZA-dC leading to activation of p53 pathway and growth arrest of cells. Furthermore, we found that the combinatorial treatment of 5-AZA-dC with three other drugs caused drug resistance. We discuss that 5-Aza-dC-induced senescence is a multi-module drug that controls cell proliferation phenotype not only by proteins but also by noncoding miRNAs. Further studies are warranted to dissect these mechanisms and establish 5-Aza-dC as an effective multi-module anticancer reagent.


Subject(s)
Antineoplastic Agents/pharmacology , Azacitidine/analogs & derivatives , Cellular Senescence/drug effects , Computational Biology , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Agents/chemistry , Azacitidine/chemistry , Azacitidine/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Decitabine , Dose-Response Relationship, Drug , Humans , Neoplasms/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Clin Rheumatol ; 36(10): 2335-2342, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28470428

ABSTRACT

The aim of this study is to identify osteoarthritis (OA)-associated differentially methylated genes in human articular chondrocytes from patients with OA. DNA methylation profiling of articular chondrocytes from OA patients, rheumatoid arthritis (RA) patients, and controls was performed, and candidate genes were chosen for validation of gene demethylation status. The mRNA expression levels of candidate genes in chondrocytes were detected by real-time quantitative PCR. Chondrocytes from OA and RA group were treated with 5-Aza-2-deoxycytidine (5-Aza), and then the mRNA expression levels were detected. Forty-five genes with significant methylation differences between OA and control group were identified. Tumor necrosis factor receptor-associated factor 1 (TRAF1), connective tissue growth factor (CTGF), and chemokine (C-X3-C motif) ligand 1(CX3CL1) genes were hypomethylated in chondrocytes of OA and RA patients, which verified by bisulfite sequencing analysis. The mRNA expression level of TRAF1 and CTGF was significantly increased in OA and RA group (p < 0.05), while the expression level of CX3CL1 was only increased in OA group (p < 0.05). For the chondrocytes from OA and RA treated with 5-Aza, the mRNA expression level of TRAF1 and CTGF was highly increased (p < 0.05). It is the first time to show that TRAF1, CTGF, and CX3CL1 genes were hypomethylated in OA chondrocytes and have a consistent correlation with mRNA expression, which suggests that epigenetic changes in the methylation status of TRAF1, CTGF, and CX3CL1 contribute to the pathology of OA.


Subject(s)
Arthritis, Rheumatoid/genetics , Cartilage, Articular/metabolism , Chemokine CX3CL1/genetics , Chondrocytes/metabolism , Connective Tissue Growth Factor/genetics , DNA Methylation , Osteoarthritis/genetics , TNF Receptor-Associated Factor 1/genetics , Aged , Aged, 80 and over , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Cells, Cultured , Chondrocytes/cytology , Decitabine , Epigenesis, Genetic , Female , Humans , Ligands , Male , Middle Aged , Osteoarthritis/metabolism , Protein Domains , RNA, Messenger/metabolism , Sample Size , Sequence Analysis, DNA , Tibia/metabolism
18.
Biotechniques ; 62(4): 157-164, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28403806

ABSTRACT

Here, we present a DNA restriction enzyme-based, fluorescent cytosine extension assay (CEA) to improve normalization and technical variation among sample-to-sample measurements. The assay includes end-labeling of parallel methylation-sensitive and methylation-insensitive DNA restriction enzyme digests along with co-purification and subsequent co-measurement of incorporated fluorescence. This non-radioactive, two-color fluorescent CEA (TCF-CEA) was shown to be a relatively rapid and accurate, with 3-fold greater precision than the one-color CEA. In addition, TCF-CEA provided an index of global DNA methylation that was sensitive to differences >5%. TCF-CEA results were highly correlated with LUminometric Methylation Assay (LUMA) results using human liver cell lines (HepG2, HepaRG, HC-04) as well as a human liver primary cell culture. Hypomethylation was observed in cells treated with the de-methylating agent 5-aza-2'-deoxycytidine. These results demonstrate that TCF-CEA provides a simple method for measuring relative degrees of global DNA methylation that could potentially be scaled up to higher-throughput formats.


Subject(s)
Biological Assay/methods , Cytosine , DNA Methylation/genetics , Azacitidine/analogs & derivatives , Azacitidine/chemistry , DNA Restriction Enzymes/genetics , Decitabine , Fluorescence , Hep G2 Cells , Humans , Liver/cytology , Liver/metabolism , Primary Cell Culture
19.
Biomed Pharmacother ; 90: 586-597, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28407579

ABSTRACT

Efficient delivery of cytidine analogues such as Azacitidine (AZA) into solid tumors constitutes a primary challenge in epigenetic therapies. We developed a di-block nano-vector based on poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) for stabilization of the conjugated AZA under physiological conditions. With equimolar drug content, our nano-conjugate could elicit a better anti-proliferative effect over free drug in breast cancer both in vitro and in vivo, through reactivation of p21 and BRCA1 to restrict cell proliferation. In addition, we applied single-molecule fluorescence tools to characterize the intracellular behavior of the AZA-PLGE-PEG nano-micelles at a finer spatiotemporal resolution. Our results suggest that the nano-micelles could effectively enrich in cancer cells and may not be limited by nucleoside transporters. Afterwards, the internalized nano-micelles exhibit pH-dependent release and resistance to active efflux. Altogether, our work describes a delivery strategy for DNA demethylating agents with nanoscale tunability, providing a cost-effective option for pharmaceutics.


Subject(s)
Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polyglactin 910/chemistry , Azacitidine/chemistry , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Carriers/chemistry , Drug Delivery Systems/methods , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Micelles , Nucleosides/chemistry
20.
Methods Mol Biol ; 1562: 269-281, 2017.
Article in English | MEDLINE | ID: mdl-28349467

ABSTRACT

Several crosslinking methods have been developed to identify interacting RNAs for proteins of interest. Here, we describe variants of the UV crosslinking and analysis of cDNA (CRAC) method that allow target identification of RNA methyltransferases on a genome-wide scale. We present a detailed protocol for the application of CRAC in human cells that stably express the protein of interest fused to a tandem affinity tag. After the introduction of a covalent link between the protein and its target RNAs, protein-RNA complexes are purified and bound RNAs trimmed, ligated to adapters, reverse transcribed, and amplified. Sequences obtained from next-generation sequencing are then mapped onto the human genome allowing the identification of possible substrates. For some RNA methyltransferases, e.g., m5C MTases, their catalytic mechanism can be exploited for chemical crosslinking approaches instead of UV based crosslinking.


Subject(s)
Methyltransferases/metabolism , RNA/genetics , RNA/metabolism , Azacitidine/chemistry , Azacitidine/metabolism , Chromatography, Affinity , Computational Biology/methods , Cytidine/analogs & derivatives , Cytidine/chemistry , Cytidine/metabolism , High-Throughput Nucleotide Sequencing , Methyltransferases/isolation & purification , RNA/chemistry , Statistics as Topic/methods , Substrate Specificity , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...