Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Nucleic Acids Res ; 49(14): 8177-8188, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34232996

ABSTRACT

The oxidative base damage, 8-oxo-7,8-dihydroguanine (8-oxoG) is a highly mutagenic lesion because replicative DNA polymerases insert adenine (A) opposite 8-oxoG. In mammalian cells, the removal of A incorporated across from 8-oxoG is mediated by the glycosylase MUTYH during base excision repair (BER). After A excision, MUTYH binds avidly to the abasic site and is thus product inhibited. We have previously reported that UV-DDB plays a non-canonical role in BER during the removal of 8-oxoG by 8-oxoG glycosylase, OGG1 and presented preliminary data that UV-DDB can also increase MUTYH activity. In this present study we examine the mechanism of how UV-DDB stimulates MUTYH. Bulk kinetic assays show that UV-DDB can stimulate the turnover rate of MUTYH excision of A across from 8-oxoG by 4-5-fold. Electrophoretic mobility shift assays and atomic force microscopy suggest transient complex formation between MUTYH and UV-DDB, which displaces MUTYH from abasic sites. Using single molecule fluorescence analysis of MUTYH bound to abasic sites, we show that UV-DDB interacts directly with MUTYH and increases the mobility and dissociation rate of MUTYH. UV-DDB decreases MUTYH half-life on abasic sites in DNA from 8800 to 590 seconds. Together these data suggest that UV-DDB facilitates productive turnover of MUTYH at abasic sites during 8-oxoG:A repair.


Subject(s)
DNA Damage/drug effects , DNA Glycosylases/genetics , Guanine/analogs & derivatives , Oxidative Stress/drug effects , Adenine/chemistry , Animals , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , DNA Replication/drug effects , DNA Replication/radiation effects , Guanine/chemistry , Guanine/pharmacology , Guanine/toxicity , Hydrocarbons, Chlorinated/pharmacology , Hydrocarbons, Chlorinated/toxicity , Mice , Oxidative Stress/radiation effects , Single Molecule Imaging
2.
Sci Rep ; 11(1): 5819, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758207

ABSTRACT

8-Oxoguanine (8-oxoG), a major oxidative base lesion, is highly accumulated in Alzheimer's disease (AD) brains during the pathogenic process. MTH1 hydrolyzes 8-oxo-dGTP to 8-oxo-dGMP, thereby avoiding 8-oxo-dG incorporation into DNA. 8-OxoG DNA glycosylase-1 (OGG1) excises 8-oxoG paired with cytosine in DNA, thereby minimizing 8-oxoG accumulation in DNA. Levels of MTH1 and OGG1 are significantly reduced in the brains of sporadic AD cases. To understand how 8-oxoG accumulation in the genome is involved in AD pathogenesis, we established an AD mouse model with knockout of Mth1 and Ogg1 genes in a 3xTg-AD background. MTH1 and OGG1 deficiency increased 8-oxoG accumulation in nuclear and, to a lesser extent, mitochondrial genomes, causing microglial activation and neuronal loss with impaired cognitive function at 4-5 months of age. Furthermore, minocycline, which inhibits microglial activation and reduces neuroinflammation, markedly decreased the nuclear accumulation of 8-oxoG in microglia, and inhibited microgliosis and neuronal loss. Gene expression profiling revealed that MTH1 and OGG1 efficiently suppress progression of AD by inducing various protective genes against AD pathogenesis initiated by Aß/Tau accumulation in 3xTg-AD brain. Our findings indicate that efficient suppression of 8-oxoG accumulation in brain genomes is a new approach for prevention and treatment of AD.


Subject(s)
Alzheimer Disease/genetics , DNA Glycosylases/genetics , Guanine/analogs & derivatives , Phosphoric Monoester Hydrolases/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , DNA Damage/drug effects , DNA Repair/drug effects , Disease Progression , Gene Expression Profiling , Guanine/metabolism , Guanine/toxicity , Humans , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Oxidative Stress/drug effects
3.
CEN Case Rep ; 8(4): 256-260, 2019 11.
Article in English | MEDLINE | ID: mdl-31154657

ABSTRACT

Acquired Fanconi syndrome has been associated with the long-term ingestion of several nucleoside analogs used to treat chronic hepatitis B virus infection. However, the nucleoside analog entecavir has not been found to cause nephrotoxicity. We report a case of entecavir-induced Fanconi syndrome. Our patient was a 73-year-old man admitted to our hospital because of renal dysfunction. He also presented with hyperaminoaciduria, renal diabetes, phosphaturia, hypophosphatemia, hypokalemia, hypouricemia, and hyperchloremic metabolic acidosis, supporting a diagnosis of Fanconi syndrome. In this case, the cause of Fanconi syndrome was most likely entecavir, which had been administered as needed depending on his renal function for 5 years. After drug discontinuation and replacement with tenofovir alafenamide fumarate therapy once a week, the patient's kidney function recovered and electrolyte anomalies partially improved. We highlight the fact that entecavir may induce severe renal dysfunction, which can cause the development of Fanconi syndrome; therefore, close monitoring of proximal tubular function is recommended during entecavir therapy.


Subject(s)
Acute Kidney Injury/chemically induced , Fanconi Syndrome/chemically induced , Guanine/analogs & derivatives , Hepatitis B, Chronic/complications , Nucleosides/toxicity , Acidosis/etiology , Acute Kidney Injury/blood , Acute Kidney Injury/complications , Acute Kidney Injury/pathology , Adenine/analogs & derivatives , Adenine/therapeutic use , Aged , Alanine , Antiviral Agents/therapeutic use , Fanconi Syndrome/blood , Fanconi Syndrome/drug therapy , Fanconi Syndrome/urine , Guanine/adverse effects , Guanine/toxicity , Hepatitis B, Chronic/drug therapy , Humans , Hypokalemia/etiology , Hypophosphatemia/etiology , Male , Nucleosides/adverse effects , Tenofovir/analogs & derivatives , Treatment Outcome , Withholding Treatment
4.
Toxicol Sci ; 160(1): 173-179, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28973694

ABSTRACT

Pregnancy is a complex physiological state, in which the metabolism of endogenous as well as exogenous agents is ostensibly altered. One exogenous agent of concern is the hepatocarcinogen aflatoxin B1 (AFB1), a foodborne fungal toxin, that requires phase I metabolic oxidation for conversion to its toxic and carcinogenic form, the AFB1-8,9-exo-epoxide. The epoxide interacts with cellular targets causing toxicity and cell death; these targets include the covalent modification of DNA leading to mutations that can initiate malignant transformation. The main detoxification pathway of the AFB1-epoxide involves phase II metabolic enzymes including the glutathione-S-transferase (GST) family. Pregnancy can modulate both phase I and II metabolism and alter the biological potency of AFB1. The present work investigated the impact of pregnancy on AFB1 exposure in mice. A single IP dose of 6 mg/kg AFB1 was administered to pregnant C57BL/6 J mice at gestation day 14 and matched non-pregnant controls. Pregnant mice accumulated 2-fold higher AFB1-N7-guanine DNA adducts in the liver when compared with nonpregnant controls 6 h post-exposure. Enhanced DNA adduct formation in pregnant animals paralleled elevated hepatic protein expression of mouse CYP1A2 and mouse homologs of human CYP3A4, phase I enzymes capable of bioactivating AFB1. Although phase II enzymes GSTA1/2 showed decreased protein expression, GSTA3, the primary enzymatic protection against the AFB1-epoxide, was unaffected at the protein level. Taken together, our results reveal that pregnancy may constitute a critical window of susceptibility for maternal health, and provide insight into the biochemical factors that could explain the underlying risks.


Subject(s)
Aflatoxin B1/analogs & derivatives , Carcinogens/toxicity , DNA Damage , Guanine/analogs & derivatives , Hepatocytes/drug effects , Liver/drug effects , Activation, Metabolic , Aflatoxin B1/metabolism , Aflatoxin B1/toxicity , Animals , Carcinogens/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , DNA Adducts/metabolism , Female , Gestational Age , Glutathione Transferase/metabolism , Guanine/metabolism , Guanine/toxicity , Hepatocytes/metabolism , Isoenzymes/metabolism , Liver/metabolism , Maternal Exposure , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
5.
Chem Res Toxicol ; 29(9): 1493-503, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27404553

ABSTRACT

The generation of chemical alkylating agents from nitrosation of glycine and bile acid conjugates in the gastrointestinal tract is hypothesized to initiate carcinogenesis. O(6)-carboxymethylguanine (O(6)-CMG) is a product of DNA alkylation derived from nitrosated glycine. Although the tendency of the structurally related adduct O(6)-methylguanine to code for the misincoporation of TTP during DNA replication is well-established, the impact of the presence of the O(6)-CMG adduct in a DNA template on the efficiency and fidelity of translesion DNA synthesis (TLS) by human DNA polymerases (Pols) has hitherto not been described. Herein, we characterize the ability of the four human TLS Pols η, ι, κ, and ζ and the replicative Pol δ to bypass O(6)-CMG in a prevalent mutational hot-spot for colon cancer. The results indicate that Pol η replicates past O(6)-CMG, incorporating dCMP or dAMP, whereas Pol κ incorporates dCMP only, and Pol ι incorporates primarily dTMP. Additionally, the subsequent extension step was carried out with high efficiency by TLS Pols η, κ, and ζ, while Pol ι was unable to extend from a terminal mismatch. These results provide a first basis of O(6)-CMG-promoted base misincorporation by Y- and B-family polymerases potentially leading to mutational signatures associated with colon cancer.


Subject(s)
DNA Adducts/chemistry , DNA Adducts/metabolism , DNA-Directed DNA Polymerase/metabolism , Guanine/analogs & derivatives , Guanine/chemistry , DNA Adducts/toxicity , DNA-Directed DNA Polymerase/chemistry , Guanine/toxicity , Humans , Mutagens/chemistry , Mutagens/metabolism , Mutagens/toxicity , Mutation , Nitrosation , Proto-Oncogene Proteins p21(ras)/genetics
6.
Chem Res Toxicol ; 29(5): 841-50, 2016 05 16.
Article in English | MEDLINE | ID: mdl-26929981

ABSTRACT

According to the electrophilic theory in toxicology, many chemical carcinogens in the environment and/or their active metabolites are electrophiles that exert their effects by forming covalent bonds with nucleophilic DNA centers. The theory of hard and soft acids and bases (HSAB), which states that a toxic electrophile reacts preferentially with a biological macromolecule that has a similar hardness or softness, clarifies the underlying chemistry involved in this critical event. Epoxides are hard electrophiles that are produced endogenously by the enzymatic oxidation of parent chemicals (e.g., alkenes and PAHs). Epoxide ring opening proceeds through a SN2-type mechanism with hard nucleophile DNA sites as the major facilitators of toxic effects. Thus, the quantitative prediction of chemical reactivity would enable a predictive assessment of the molecular potential to exert electrophile-mediated toxicity. In this study, we calculated the activation energies for reactions between epoxides and the guanine N7 site for a diverse set of epoxides, including aliphatic epoxides, substituted styrene oxides, and PAH epoxides, using a state-of-the-art density functional theory (DFT) method. It is worth noting that these activation energies for diverse epoxides can be further predicted by quantum chemically calculated nucleophilic indices from HSAB theory, which is a less computationally demanding method than the exacting procedure for locating the transition state. More importantly, the good qualitative/quantitative correlations between the chemical reactivity of epoxides and their bioactivity suggest that the developed model based on HSAB theory may aid in the predictive hazard evaluation of epoxides, enabling the early identification of mutagenicity/carcinogenicity-relevant SN2 reactivity.


Subject(s)
Acids/chemistry , Alkalies/chemistry , Epoxy Compounds/chemistry , Guanine/toxicity , Guanine/chemistry , Models, Chemical
7.
J Biol Chem ; 291(10): 5309-19, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26733197

ABSTRACT

The well known biomarker of oxidative stress, 8-oxo-7,8-dihydroguanine, is more susceptible to further oxidation than the parent guanine base and can be oxidatively transformed to the genotoxic spiroiminodihydantoin (Sp) and 5-guanidinohydantoin (Gh) lesions. Incubation of 135-mer duplexes with single Sp or Gh lesions in human cell extracts yields a characteristic nucleotide excision repair (NER)-induced ladder of short dual incision oligonucleotide fragments in addition to base excision repair (BER) incision products. The ladders were not observed when NER was inhibited either by mouse monoclonal antibody (5F12) to human XPA or in XPC(-/-) fibroblast cell extracts. However, normal NER activity appeared when the XPC(-/-) cell extracts were complemented with XPC-RAD23B proteins. The Sp and Gh lesions are excellent substrates of both BER and NER. In contrast, 5-guanidino-4-nitroimidazole, a product of the oxidation of guanine in DNA by peroxynitrite, is an excellent substrate of BER only. In the case of mouse embryonic fibroblasts, BER of the Sp lesion is strongly reduced in NEIL1(-/-) relative to NEIL1(+/+) extracts. In summary, in human cell extracts, BER and NER activities co-exist and excise Gh and Sp DNA lesions, suggesting that the relative NER/BER product ratios may depend on competitive BER and NER protein binding to these lesions.


Subject(s)
DNA Repair , Guanine/analogs & derivatives , Oxidative Stress , Animals , Cell Line , Cells , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Guanine/metabolism , Guanine/toxicity , HeLa Cells , Humans , Mice
8.
Mutat Res ; 779: 24-32, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26125998

ABSTRACT

Hydroxyl radical (OH) among reactive oxygen species cause damage to nucleobases with thymine being the most susceptible, whilst in contrast, the singlet oxygen ((1)02) targets only guanine bases. The high GC content of mycobacterial genomes predisposes these organisms to oxidative damage of guanine. The exposure of cellular DNA to OH and one-electron oxidants results in the formation of two main degradation products, the pro-mutagenic 8-oxo-7,8-dihydroguanine (8-oxoGua) and the cytotoxic 2,6-diamino-4-hydroxy-5-formamidopyrimidine (FapyGua). These lesions are repaired through the base excision repair (BER) pathway and we previously, demonstrated a combinatorial role for the mycobacterial Endonuclease III (Nth) and the Nei family of DNA glycosylases in mutagenesis. In addition, the formamidopyrimidine (Fpg/MutM) and MutY DNA glycosylases have also been implicated in mutation avoidance and BER in mycobacteria. In this study, we further investigate the combined role of MutY and the Fpg/Nei DNA glycosylases in Mycobacterium smegmatis and demonstrate that deletion of mutY resulted in enhanced sensitivity to oxidative stress, an effect which was not exacerbated in Δfpg1 Δfpg2 or Δnei1 Δnei2 double mutant backgrounds. However, combinatorial loss of the mutY, fpg1 and fpg2 genes resulted in a significant increase in mutation rates suggesting interplay between these enzymes. Consistent with this, there was a significant increase in C → A mutations with a corresponding change in cell morphology of rifampicin resistant mutants in the Δfpg1 Δfpg2 ΔmutY deletion mutant. In contrast, deletion of mutY together with the nei homologues did not result in any growth/survival defects or changes in mutation rates. Taken together these data indicate that the mycobacterial mutY, in combination with the Fpg DNA N-glycosylases, plays an important role in controlling mutagenesis under oxidative stress.


Subject(s)
DNA Glycosylases/genetics , DNA Repair/genetics , DNA-Formamidopyrimidine Glycosylase/genetics , Mycobacterium smegmatis/enzymology , DNA Glycosylases/metabolism , DNA-Formamidopyrimidine Glycosylase/metabolism , Genome, Bacterial/drug effects , Guanine/analogs & derivatives , Guanine/metabolism , Guanine/toxicity , Hydroxyl Radical/toxicity , Mutagenesis/genetics , Mutagens/metabolism , Mutagens/toxicity , Mutation , Oxidative Stress/drug effects , Oxidative Stress/genetics , Pyrimidines/metabolism , Pyrimidines/toxicity
9.
Dent Mater ; 31(10): 1159-68, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26187530

ABSTRACT

OBJECTIVE: Released components of oral biomaterials can leach into the oral cavity and may subsequently reach the gastrointestinal tract. Camphorquinone (CQ) is the most common used photoinitiator in resinous restorative materials and is often combined with the co-initiator N,N-dimethyl-p-toluidine (DMT). It has been shown that CQ exerts cytotoxic effects, at least partially due to the generation of reactive oxygen species (ROS). Objective of this study was to examine the cytotoxic and genotoxic potential of CQ in human oral keratinocytes (OKF6/TERT2) and immortalized epithelial colorectal adenocarcinoma cells (Caco-2). Furthermore, the effects of visible-light irradiation and the co-initiator DMT were investigated as well as the generation of ROS, the potential protective effect of glutathione (GSH) and a recovery period of CQ-treated Caco-2 cells. METHODS: The alkaline comet assay was used to determine DNA damage. Additionally, an enzyme modified comet assay was applied, which detects 7,8-dihydro-8-oxoguanine (8-oxoguanine), a reliable marker for oxidative stress. RESULTS: Our data revealed that high concentrations of CQ induced DNA lesions in OKF6/TERT2 cells. This DNA damage is at least partly caused by the generation of 8-oxoguanine. In addition, CQ and DMT increased ROS formation and induced DNA damage in Caco-2 cells. CQ-treatment resulted in generation of 8-oxoguanine. The antioxidant GSH efficiently prevented CQ-associated DNA damage. Furthermore, a recovery following CQ-treatment significantly reduced DNA damage. SIGNIFICANCE: We conclude that CQ-induced DNA damage is caused by oxidative stress in oral and intestinal cells. These lesions can be prevented and possibly repaired by GSH-treatment and recovery of cells after the photoinitiator is removed from cultures.


Subject(s)
Antioxidants/pharmacology , Caco-2 Cells/drug effects , Camphor/analogs & derivatives , Glutathione/pharmacology , Keratinocytes/drug effects , Toluidines/toxicity , Camphor/toxicity , Comet Assay , DNA Damage , Guanine/analogs & derivatives , Guanine/toxicity , Humans , In Vitro Techniques , Mouth Mucosa/cytology , Oxidative Stress , Reactive Oxygen Species/toxicity
10.
BMC Cancer ; 14: 508, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25012241

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) patients with interstitial lung disease (ILD) need to be approached carefully given the high incidence of pulmonary toxicity. Pemetrexed (PEM) is the key drug for the treatment of NSCLC. However, its safety, especially with respect to the exacerbation of ILD, and efficacy in NSCLC patients with ILD have yet to be established. METHOD: We investigated the safety and efficacy of PEM monotherapy in NSCLC patients with or without idiopathic interstitial pneumonia (IIPs). The medical charts of these patients were retrospectively reviewed. RESULTS: Twenty-five patients diagnosed as having IIPs (IIPs group) and 88 patients without ILD (non-ILD group) were treated with PEM monotherapy at Juntendo University Hospital between 2009 and 2013. In the IIPs group, 12 patients were found to have usual interstitial pneumonitis (UIP) on chest computed tomography (CT) (UIP group) and the other 13 patients showed a non-UIP pattern on chest CT (non-UIP IIPs group). Three patients in the IIPs group (2 in the UIP group and 1 in the non-UIP IIPs group) and 1 in the non-ILD group developed pulmonary toxicity during treatment (3.5% overall, 12.0% in the IIPs group versus 1.1% in the non-ILD group). Moreover, all 3 patients in the IIPs group died of pulmonary toxicity. Overall survival tended to be longer in the non-ILD group than in the IIPs group (p = 0.08). Multivariate analyses demonstrated that IIPs was the only significant independent risk factor for PEM-related pulmonary toxicity. CONCLUSION: We found that the incidence of PEM-related pulmonary toxicity was significantly higher amongst NSCLC patients with IIPs than among those without IIPs. Particular care must be taken when administering PEM to treat NSCLC patients with IIPs.


Subject(s)
Antineoplastic Agents/toxicity , Carcinoma, Non-Small-Cell Lung/drug therapy , Glutamates/toxicity , Guanine/analogs & derivatives , Idiopathic Interstitial Pneumonias/complications , Idiopathic Interstitial Pneumonias/mortality , Lung Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Female , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/toxicity , Humans , Male , Middle Aged , Pemetrexed , Survival Analysis
11.
DNA Repair (Amst) ; 21: 55-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25037770

ABSTRACT

Functional DNA mismatch repair (MMR) is essential for maintaining the fidelity of DNA replication and genetic stability. In hematopoiesis, loss of MMR results in methylating agent resistance and a hematopoietic stem cell (HSC) repopulation defect. Additionally MMR failure is associated with a variety of human malignancies, notably Lynch syndrome. We focus on the 5'→3' exonuclease Exo1, the primary enzyme excising the nicked strand during MMR, preceding polymerase synthesis. We found that nuclease dead Exo1 mutant cells are sensitive to the O6-methylguanine alkylating agent temozolomide when given with the MGMT inactivator, O6benzylguanine (BG). Additionally we used an MMR reporter plasmid to verify that Exo1(mut) MEFs were able to repair G:T base mismatches in vitro. We showed that unlike other MMR deficient mouse models, Exo1(mut) mouse HSC did not gain a competitive survival advantage post temozolomide/BG treatment in vivo. To determine potential nucleases implicated in MMR in the absence of Exo1 nuclease activity, but in the presence of the inactive protein, we performed gene expression analyses of several mammalian nucleases in WT and Exo1(mut) MEFs before and after temozolomide treatment and identified upregulation of Artemis, Fan1, and Mre11. Partial shRNA mediated silencing of each of these in Exo1(mut) cells resulted in decreased MMR capacity and increased resistance to temozolomide/BG. We propose that nuclease function is required for fully functional MMR, but a portfolio of nucleases is able to compensate for loss of Exo1 nuclease activity to maintain proficiency.


Subject(s)
DNA Mismatch Repair , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Endonucleases/metabolism , Exodeoxyribonucleases/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/toxicity , Endonucleases/genetics , Exodeoxyribonucleases/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Guanine/analogs & derivatives , Guanine/toxicity , MRE11 Homologue Protein , Mice , Mice, Inbred C57BL , Multifunctional Enzymes , Nuclear Proteins/genetics , Temozolomide , Up-Regulation
12.
Mol Med Rep ; 10(2): 931-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24840891

ABSTRACT

Currently, chemotherapy and targeted therapies share the principal limitation of the emergence of drug resistance, which prevents these strategies from having lasting clinical benefits. The combination of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) with concurrent chemotherapy has been proposed as one strategy to overcome acquired resistance to EGFR-TKIs. The purpose of the present study was to investigate the combined effects of gefitinib and pemetrexed on EGFR-TKI-sensitive and EGFR­TKI­resistant human non-small cell lung cancer (NSCLC) cell lines. The antiproliferative effects of gefitinib and pemetrexed, alone and in combination, on the growth of NSCLC cell lines, were assessed using an MTT assay. The cytotoxic interaction between the two drugs was evaluated in vitro using the combination index (CI) method. Cell cycle distribution and apoptosis were analyzed by flow cytometry and alterations in signaling pathways were determined by western blot analysis. In the present study, it was identified that when cells were concurrently exposed to pemetrexed and gefitinib, cytotoxic synergism was present in the gefitinib-resistant PC9/GR human NSCLC cell line and antagonistic interactions were observed in the gefitinib-sensitive PC9 cell line. Synergism was associated with a combination of cell cycle effects of the different agents. In addition, the combination of pemetrexed and gefitinib decreased the levels of phosphorylated AKT, phosphorylated extracellular-signal-regulated kinase and B-cell lymphoma 2 as compared with those in the control. By contrast, antagonism was associated with gefitinib-induced G0/G1-phase blockade of gefitinib-sensitive cells, which interfered with the cell cycle-specific cytotoxicity of chemotherapy. The combination of pemetrexed and gefitinib generated synergistic effects in gefitinib-acquired resistant cells and antagonistic effects in gefitinib-sensitive cells, suggesting that EGFR-TKIs combined with pemetrexed may be a beneficial treatment strategy for NSCLC patients with acquired resistance to EGFR-TKIs.


Subject(s)
Antineoplastic Agents/toxicity , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Glutamates/toxicity , Guanine/analogs & derivatives , Quinazolines/toxicity , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Gefitinib , Guanine/toxicity , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Pemetrexed , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
13.
Yonsei Med J ; 54(4): 854-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23709418

ABSTRACT

PURPOSE: The main objective of this study was to evaluate the association between polymorphisms of the target genes of pemetrexed and clinical outcomes in non-small cell lung cancer (NSCLC) patients treated with pemetrexed. MATERIALS AND METHODS: We assessed polymorphisms at 8 sites in 4 genes [thymidylate synthase (TS), dihydrofolate reductase (DHFR; 1610, 680, 317, intron 1), methylenetetrahydrofolate reductase (MTHFR; 677, 1298), glycinamide ribonucleotide formyl transferase (GARFT; 2255)] associated with pemetrexed metabolism using polymerase chain reaction, gene scanning, and restriction fragment length polymorphism analysis in 90 patients with adenocarcinoma of the lung. RESULTS: Survival was significantly longer with pemetrexed in patients with TS 3RGCC/3RGCC or 3RGGC/3RGGC compared with the other groups (PFS; 5.2 months vs. 3.7 months, p=0.03: OS; 31.8 months vs. 18.5 months, p=0.001). Patients with DHFR 680CC experienced fatigue more frequently (50% vs. 8.6%, p=0.008). Polymorphisms of MTHFR and GARFT were not significantly associated with clinical outcomes of pemetrexed. CONCLUSION: The TS genotype was associated with survival and one DHFR polymorphism was associated with fatigue in NSCLC patients treated with pemetrexed. Further large prospective studies are required to identify other biomarkers that affect patients being treated with pemetrexed for adenocarcinoma of the lung.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Antimetabolites, Antineoplastic/therapeutic use , Glutamates/therapeutic use , Guanine/analogs & derivatives , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Polymorphism, Single Nucleotide , Adenocarcinoma/mortality , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/toxicity , Female , Glutamates/pharmacology , Glutamates/toxicity , Guanine/pharmacology , Guanine/therapeutic use , Guanine/toxicity , Humans , Lung Neoplasms/mortality , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Pemetrexed , Pharmacogenetics , Phosphoribosylglycinamide Formyltransferase/genetics , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/genetics
14.
Clin Oral Investig ; 17(3): 1009-15, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22777392

ABSTRACT

OBJECTIVES: The aim of this study was to evaluate the physical properties and cytotoxicity of a novel root-end filling material (EPC) which is made from epoxy resin and Portland cement as a mineral trioxide aggregate (MTA) substitute. MATERIALS AND METHODS: EPC, developed as a root-end filling material, was compared with MTA and a mixture of AH Plus sealer and MTA (AMTA) with regard to the setting time, radio-opacity, and microleakage. Setting times were evaluated using Vicat apparatus. Digital radiographs were taken to evaluate the aluminium equivalent radio-opacity using an aluminium step wedge. Extracted single-rooted teeth were used for leakage test using methylene blue dye. After canal shaping and obturation, the apical 3-mm root was resected, and a root-end cavity with a depth of 3 mm was prepared. The root-end cavities were filled with MTA, AMTA, and EPC for 15 specimens in each of three groups. After setting in humid conditions for 24 h, the specimens were tested for apical leakage. For evaluation of the biocompatibility of EPC, cell (human gingival fibroblast) viability was compared for MTA and Portland cement by MTT assay, and cell morphological changes were compared for MTA and AH Plus by fluorescence microscopy using DAPI and F-actin staining. The setting time, radio-opacity, and microleakage were compared using one-way ANOVA and Scheffe's post hoc comparison, and the cytotoxicity was compared using the nonparametric Kruskal-Wallis rank sum test. Statistical significance was set at 95%. RESULTS: EPC had a shorter setting time and less microleakage compared with MTA (p < 0.05). EPC showed 5-mm aluminium thickness radio-opacity and similar biocompatibility to MTA. CONCLUSIONS: Under the conditions of this study, EPC, a novel composite made from a mixture of epoxy resin and Portland cement, was found to be a useful material for root-end filling, with favourable radio-opacity, short setting time, low microleakage, and clinically acceptable low cytotoxicity. CLINICAL RELEVANCE: The novel root-end filling material would be a potentially useful material for a surgical endodontic procedure with favourable properties.


Subject(s)
Dental Cements , Epoxy Resins/chemistry , Root Canal Filling Materials/chemistry , Biocompatible Materials/chemistry , Cell Survival/drug effects , Dental Leakage , Epoxy Resins/toxicity , Gingiva/cytology , Gingiva/drug effects , Glutamates/chemistry , Glutamates/toxicity , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/toxicity , Humans , Materials Testing , Pemetrexed , Retrograde Obturation , Root Canal Filling Materials/toxicity
15.
Biochemistry (Mosc) ; 77(3): 270-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22803944

ABSTRACT

Trinucleotide repeat expansion provides a molecular basis for several devastating neurodegenerative diseases. In particular, expansion of a CAG run in the human HTT gene causes Huntington's disease. One of the main reasons for triplet repeat expansion in somatic cells is base excision repair (BER), involving damaged base excision and repair DNA synthesis that may be accompanied by expansion of the repaired strand due to formation of noncanonical DNA structures. We have analyzed the kinetics of excision of a ubiquitously found oxidized purine base, 8-oxoguanine (oxoG), by DNA glycosylase OGG1 from the substrates containing a CAG run flanked by AT-rich sequences. The values of k(2) rate constant for the removal of oxoG from triplets in the middle of the run were higher than for oxoG at the flanks of the run. The value of k(3) rate constant dropped starting from the third CAG-triplet in the run and remained stable until the 3'-terminal triplet, where it decreased even more. In nuclear extracts, the profile of oxoG removal rate along the run resembled the profile of k(2) constant, suggesting that the reaction rate in the extracts is limited by base excision. The fully reconstituted BER was efficient with all substrates unless oxoG was near the 3'-flank of the run, interfering with the initiation of the repair. DNA polymerase ß was able to perform a strand-displacement DNA synthesis, which may be important for CAG run expansion initiated by BER.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Guanine/analogs & derivatives , Trinucleotide Repeats/drug effects , Cell Line , DNA Glycosylases/genetics , Guanine/toxicity , Humans
16.
Hum Mol Genet ; 21(7): 1470-80, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22171072

ABSTRACT

Pemetrexed, approved for the treatment of non-small cell lung cancer and malignant mesothelioma, has adverse effects including neutropenia, leucopenia, thrombocytopenia, anemia, fatigue and nausea. The results we report here represent the first genome-wide study aimed at identifying genetic predictors of pemetrexed response. We utilized expression quantitative trait loci (eQTLs) mapping combined with drug-induced cytotoxicity data to gain mechanistic insights into the observed genetic associations with pemetrexed susceptibility. We found that CTTN and ZMAT3 expression signature explained >30% of the pemetrexed susceptibility phenotype variation for pemetrexed in the discovery population. Replication using PCR and a semi-high-throughput, scalable assay system confirmed the initial discovery results in an independent set of samples derived from the same ancestry. Furthermore, functional validation in both germline and tumor cells demonstrates a decrease in cell survival following knockdown of CTTN or ZMAT3. In addition to our particular findings on genetic and gene expression predictors of susceptibility phenotype for pemetrexed, the work presented here will be valuable to the robust discovery and validation of genetic determinants and gene expression signatures of various chemotherapeutic susceptibilities.


Subject(s)
Antineoplastic Agents/toxicity , Carrier Proteins/genetics , Cortactin/genetics , Glutamates/toxicity , Guanine/analogs & derivatives , Nuclear Proteins/genetics , Quantitative Trait Loci , Carrier Proteins/metabolism , Cell Line, Tumor , Cortactin/metabolism , Gene Expression , Genome-Wide Association Study , Guanine/toxicity , Humans , Linear Models , Lymphocytes/drug effects , Lymphocytes/metabolism , Nuclear Proteins/metabolism , Pemetrexed , Polymorphism, Single Nucleotide , RNA-Binding Proteins
17.
Rev Mal Respir ; 28(1): 97-100, 2011 Jan.
Article in French | MEDLINE | ID: mdl-21277483

ABSTRACT

INTRODUCTION: Pneumocystis pneumonia is a life-threatening infection in patients undergoing chemotherapy for solid malignancies. CASE REPORT: A 49-year-old man developed gradually increasing dyspnoea while receiving pemetrexed as a third line treatment for an adenocarcinoma of the lung. The diagnosis of pneumocystis pneumonia was based on ground-glass opacities on the thoracic CT scan and alveolar lavage revealing occasional cysts of Pneumocystis jiroveci in the context of recent lymphopenia developing during chemotherapy. Treatment with cotrimoxazole for three weeks was only partially successful due to progression of the tumour. CONCLUSIONS: Pneumocystis pneumonia should be considered in cancer patients receiving antifolate drugs and presenting with increasing dyspnoea. It is important to identify a high-risk population among patients undergoing chemotherapy because of the significant morbidity and mortality and in order to administer effective prophylactic agents.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/toxicity , Glutamates/toxicity , Guanine/analogs & derivatives , Lung Neoplasms/drug therapy , Opportunistic Infections/diagnosis , Pneumocystis carinii , Pneumonia, Pneumocystis/diagnosis , Antifungal Agents/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Bronchoalveolar Lavage Fluid/microbiology , Disease Progression , Follow-Up Studies , Glutamates/therapeutic use , Guanine/therapeutic use , Guanine/toxicity , Humans , Infusions, Intravenous , Male , Middle Aged , Pemetrexed , Pneumonia, Pneumocystis/drug therapy , Retreatment , Tomography, X-Ray Computed , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use
18.
Chemotherapy ; 56(1): 54-9, 2010.
Article in English | MEDLINE | ID: mdl-20215748

ABSTRACT

BACKGROUND/AIMS: 9-[2-(phosphonomethoxy)ethyl]guanine (PMEG) is one of the most active antiproliferative compounds in a series of acyclic nucleoside phosphonates and is active in intraperitoneal P388 tumors in mice. METHODS: We synthesized octadecyloxyethyl (ODE) and hexadecyloxypropyl esters of PMEG and compared their antiproliferative activity with unmodified PMEG in primary human fibroblasts and CaSki, Me-180 and HeLa human cervical cancer cell lines in vitro. RESULTS: ODE-PMEG had excellent antiproliferative activity in vitro in this panel of human cervical cancers. We compared the effects of ODE-PMEG and ODE-cidofovir (ODE-CDV) in a solid tumor model using Me-180 human cervical cancer cell lines in athymic nude mice. Intratumoral injection of 25 microg of ODE-PMEG or 100 microg of ODE-CDV daily for 21 days followed by observation for 20-35 days resulted in near-complete disappearance of measurable cervical cancers. CONCLUSION: ODE-PMEG may be suitable for local or topical treatment of cervical dysplasia.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Guanine/analogs & derivatives , Organophosphonates/therapeutic use , Organophosphorus Compounds/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Adenine/chemistry , Adenine/therapeutic use , Adenine/toxicity , Animals , Antineoplastic Agents/toxicity , Female , Guanine/therapeutic use , Guanine/toxicity , HeLa Cells , Humans , Mice , Mice, Nude , Organophosphonates/chemistry , Organophosphonates/toxicity , Organophosphorus Compounds/toxicity , Tumor Cells, Cultured
19.
Curr Drug Targets ; 11(1): 48-57, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19839928

ABSTRACT

Pemetrexed is a multi-targeted anti metabolite that inhibits several key folate-dependent enzymes in the thymidine and purine biosynthetic pathways, including thymidylate synthase. It is currently approved for use in patients with non-small cell lung cancer and malignant mesothelioma. The sporadic and unpredictable occurrence of haematological toxicities of pemetrexed leading to potentially life threatening complications during the early developmental phase, prompted urgent need to identify potential predictive factors for haematological toxicities from pemetrexed. There is a well established association between elevated plasma homocysteine concentration, which is indicative of impaired functional folate status, and increased risk of haematological toxicity from pemetrexed. The decrease in incidence of toxicity after vitamin supplementation confirms the importance of functional folate status as a predictor for haematological toxicity. We review other factors that have a documented impact on haematological toxicity, including pemetrexed schedule, and pharmacokinetic parameters that are indicative of the extent of drug exposure. Further potential factors are explored in this review, such as the genotype of the pemetrexed metabolising enzymes and varying incidences of polymorphism of these genotypes in different ethnic groups that may account for the ethnic differences in neutropenic response to pemetrexed.


Subject(s)
Glutamates/toxicity , Guanine/analogs & derivatives , Hematologic Diseases/chemically induced , Biomarkers/blood , Biomarkers/metabolism , Drug-Related Side Effects and Adverse Reactions/blood , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/metabolism , Glutamates/administration & dosage , Glutamates/pharmacology , Guanine/administration & dosage , Guanine/pharmacology , Guanine/toxicity , Humans , Pemetrexed
20.
Environ Mol Mutagen ; 51(3): 236-42, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19844954

ABSTRACT

The aim of this work was to determine the kinetics of micronucleus production because of an increase in O(6)-chloroethyl guanine (O6-ChlEt-G) DNA lesions in murine bone marrow cells in vivo. We increased the frequency of O6-ChlEt-G lesions by pretreatment with an inhibitor of O(6)-methylguanine-DNA methyltransferase (MGMT), O(6)-benzylguanine (O6BG), and subsequent treatment with bis-chloroethylnitrosourea (BCNU). The kinetics of micronucleated-polychromatic erythrocyte (MN-PCE) induction was established by scoring the frequency of MN-PCEs per 2000 PCEs in peripheral blood at 8-hr intervals from immediately prior to treatment to 72-hr post-treatment. We examined groups of five mice treated with (i) dimethylsulfoxide (DMSO), (ii) O6BG in DMSO, (iii) BCNU, or (iv) O6BG in DMSO plus BCNU. The data indicate that O6BG pretreatment causes: (i) ían increase in MN-PCEs induced by BCNU, (ii) a delay in the time of maximal MN-PCE induction produced by the different BCNU doses, and (iii) an increase in cytotoxicity. These data confirm that O6-ChlEt-G is a lesion involved in DNA break induction and in the subsequent production of micronuclei, and also that these lesions seem to be stoichiometrically reduced by MGMT. These data also show that induction of MN-PCEs by BCNU is delayed by pretreatment with O6BG for more than 6 hr, perhaps due to the time required for repair of crosslinks derived from O6-ChlEt-G and/or for DNA duplication, which is required for adduct transformation into crosslinks.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Carmustine/toxicity , DNA Damage , Guanine/analogs & derivatives , Micronuclei, Chromosome-Defective/chemically induced , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/toxicity , Erythrocytes/drug effects , Erythrocytes/metabolism , Guanine/metabolism , Guanine/toxicity , Kinetics , Male , Mice , Mice, Inbred BALB C , Micronuclei, Chromosome-Defective/drug effects , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...