Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Redox Biol ; 69: 102977, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38056311

ABSTRACT

Ref-1/APE1 (Redox Effector/Apurinic Endonuclease 1) is a multifunctional enzyme that serves as a redox factor for several transcription factors (TFs), e.g., NF-kB, HIF-1α, which in an oxidized state fail to bind DNA. Conversion of these TFs to a reduced state serves to regulate various biological responses such as cell growth, inflammation, and cellular metabolism. The redox activity involves a thiol exchange reaction for which Cys65 (C65) serves as the nucleophile. Using CRISPR editing in human pancreatic ductal adenocarcinoma (PDAC) cells, we changed C65 to Ala (C65A) in Ref-1 to evaluate alteration of Ref-1 redox dynamics as well as chronic loss of Ref-1 redox activity on cell signaling pathways, specifically those regulated by NF-kB and HIF-1α. The redox activity of Ref-1 requires partial unfolding to expose C65, which is buried in the folded structure. Labeling of Ref-1 with polyethylene glycol-maleimide (PEGm) provides a readout of reduced Cys residues in Ref-1 and thereby an assessment of partial unfolding in Ref-1. In comparing Ref-1WT vs Ref-1C65A cell lines, we found an altered distribution of oxidized versus reduced states of Ref-1. Accordingly, activation of NF-kB and HIF-1α in Ref-1C65A lines was significantly lower compared to Ref-1WT lines. The bioinformatic data revealed significant downregulation of metabolic pathways including OXPHOS in Ref-1C65A expressing clones compared to Ref-1WT line. Ref-1C65A also demonstrated reduced cell proliferation and use of tricarboxylic acid (TCA) substrates compared to Ref-1WT lines. A subcutaneous as well as PDAC orthotopic in vivo model demonstrated a significant reduction in tumor size, weight, and growth in the Ref-1C65A lines compared to the Ref-1WT lines. Moreover, mice implanted with Ref-1C65A redox deficient cells demonstrate significantly reduced metastatic burden to liver and lung compared to mice implanted with Ref-1 redox proficient cells. These results from the current study provide direct evidence that the chronic absence of Cys65 in Ref-1 results in redox inactivity of the protein in human PDAC cells, and subsequent biological results confirm a critical involvement of Ref-1 redox signaling and tumorigenic phenotype.


Subject(s)
NF-kappa B , Pancreatic Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Cell Proliferation , Cysteine/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , NF-kappa B/metabolism , Oxidation-Reduction , Pancreatic Neoplasms/pathology , Signal Transduction
2.
Nat Commun ; 12(1): 6543, 2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34764304

ABSTRACT

The Moon presently has no dynamo, but magnetic fields have been detected over numerous portions of its crust. Most of these regions are located antipodal to large basins, leading to the hypothesis that lunar rock ejected during basin-forming impacts accumulated at the basin antipode and recorded the ambient magnetic field. However, a major problem with this hypothesis is that lunar materials have low iron content and cannot become strongly magnetized. Here we simulate oblique impacts of 100-km-diameter impactors at high resolution and show that an ~700 m thick deposit of potentially iron-rich impactor material accumulates at the basin antipode. The material is shock-heated above the Curie temperature and therefore may efficiently record the ambient magnetic field after deposition. These results explain a substantial fraction of the Moon's crustal magnetism, and are consistent with a dynamo field strength of at least several tens of microtesla during the basin-forming epoch.

3.
Redox Biol ; 2: 485-94, 2014.
Article in English | MEDLINE | ID: mdl-24624338

ABSTRACT

The effectiveness of current treatment for age related macular degeneration (AMD) by targeting one molecule is limited due to its multifactorial nature and heterogeneous pathologies. Treatment strategy to target multiple signaling pathways or pathological components in AMD pathogenesis is under investigation for better clinical outcome. Inhibition of the redox function of apurinic endonuclease 1/redox factor-1 (APE1) was found to suppress endothelial angiogenesis and promote neuronal cell recovery, thereby may serve as a potential treatment for AMD. In the current study, we for the first time have found that a specific inhibitor of APE1 redox function by a small molecule compound E3330 regulates retinal pigment epithelium (RPEs) cell response to oxidative stress. E3330 significantly blocked sub-lethal doses of oxidized low density lipoprotein (oxLDL) induced proliferation decline and senescence advancement of RPEs. At the same time, E3330 remarkably decreased the accumulation of intracellular reactive oxygen species (ROS) and down-regulated the productions of monocyte chemoattractant protein-1 (MCP-1) and vascular endothelial growth factor (VEGF), as well as attenuated the level of nuclear factor-κB (NF-κB) p65 in RPEs. A panel of stress and toxicity responsive transcription factors that were significantly upregulated by oxLDL was restored by E3330, including Nrf2/Nrf1, p53, NF-κB, HIF1, CBF/NF-Y/YY1, and MTF-1. Further, a single intravitreal injection of E3330 effectively reduced the progression of laser-induced choroidal neovascularization (CNV) in mouse eyes. These data revealed that E3330 effectively rescued RPEs from oxidative stress induced senescence and dysfunctions in multiple aspects in vitro, and attenuated laser-induced damages to RPE-Bruch׳s membrane complex in vivo. Together with its previously established anti-angiogenic and neuroprotection benefits, E3330 is implicated for potential use for AMD treatment.


Subject(s)
Benzoquinones/administration & dosage , Choroidal Neovascularization/drug therapy , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Neuroprotective Agents/administration & dosage , Oxidative Stress/drug effects , Propionates/administration & dosage , Retinal Pigment Epithelium/metabolism , Animals , Cellular Senescence/drug effects , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Intravitreal Injections , Mice , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology
4.
Neurotoxicology ; 32(1): 140-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20932997

ABSTRACT

Peripheral neuropathy is a major side effect following treatment with the cancer chemotherapeutic drug paclitaxel. Whether paclitaxel-induced peripheral neuropathy is secondary to altered function of small diameter sensory neurons remains controversial. To ascertain whether the function of the small diameter sensory neurons was altered following systemic administration of paclitaxel, we injected male Sprague Dawley rats with 1mg/kg paclitaxel every other day for a total of four doses and examined vasodilatation in the hindpaw at day 14 as an indirect measure of calcitonin gene related peptide (CGRP) release. In paclitaxel-treated rats, the vasodilatation induced by either intradermal injection of capsaicin into the hindpaw or electrical stimulation of the sciatic nerve was significantly attenuated in comparison to vehicle-injected animals. Paclitaxel treatment, however, did not affect direct vasodilatation induced by intradermal injection of methacholine or CGRP, demonstrating that the blood vessels' ability to dilate was intact. Paclitaxel treatment did not alter the compound action potentials or conduction velocity of C-fibers. The stimulated release of CGRP from the central terminals in the spinal cord was not altered in paclitaxel-injected animals. These results suggest that paclitaxel affects the peripheral endings of sensory neurons to alter transmitter release, and this may contribute to the symptoms seen in neuropathy.


Subject(s)
Hindlimb/blood supply , Hindlimb/drug effects , Paclitaxel/toxicity , Sensory Receptor Cells/drug effects , Vasodilation/drug effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Hindlimb/physiology , Male , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology , Vasodilation/physiology
5.
Mutat Res ; 643(1-2): 54-63, 2008 Aug 25.
Article in English | MEDLINE | ID: mdl-18579163

ABSTRACT

Human apurinic/apyrimidinic endonuclease (hApe1) encodes two important functional activities: an essential base excision repair (BER) activity and a redox activity that regulates expression of a number of genes through reduction of their transcription factors, AP-1, NFkappaB, HIF-1alpha, CREB, p53 and others. The BER function is highly conserved from prokaryotes (E. coli exonuclease III) to humans (hApe1). Here, we provide evidence supporting a redox function unique to mammalian Apes. An evolutionary analysis of Ape sequences reveals that, of the 7 Cys residues, Cys 93, 99, 208, 296, and 310 are conserved in both mammalian and non-mammalian vertebrate Apes, while Cys 65 is unique to mammalian Apes. In the zebrafish Ape (zApe), selected as the vertebrate sequence most distant from human, the residue equivalent to Cys 65 is Thr 58. The wild-type zApe enzyme was tested for redox activity in both in vitro EMSA and transactivation assays and found to be inactive, similar to C65A hApe1. Substitution of Thr 58 with Cys in zApe, however, resulted in a redox active enzyme, suggesting that a Cys residue in this position is indeed critical for redox function. In order to further probe differences between redox active and inactive enzymes, we have determined the crystal structures of vertebrate redox inactive enzymes, the C65A human Ape1 enzyme and the zApe enzyme at 1.9 and 2.3A, respectively. Our results provide new insights on the redox function and highlight a dramatic gain-of-function activity for Ape1 in mammals not found in non-mammalian vertebrates or lower organisms.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Evolution, Molecular , Zebrafish/metabolism , Amino Acid Sequence , Animals , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Electrophoretic Mobility Shift Assay , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Sequence Homology, Amino Acid , Transcriptional Activation , Zebrafish/genetics
6.
Nucleic Acids Res ; 33(9): 2859-67, 2005.
Article in English | MEDLINE | ID: mdl-15905475

ABSTRACT

Previous studies indicate that overexpression of N-methylpurine DNA glycosylase (MPG) dramatically sensitizes cells to alkylating agent-induced cytotoxicity. We recently demonstrated that this sensitivity is preceded by an increased production of AP sites and strand breaks, confirming that overexpression of MPG disrupts normal base excision repair and causes cell death through overproduction of toxic repair intermediates. Here we establish through site-directed mutagenesis that MPG-induced sensitivity to alkylation is dependent on enzyme glycosylase activity. However, in contrast to the sensitivity seen to heterogeneous alkylating agents, MPG overexpression generates no cellular sensitivity to MeOSO2(CH2)2-lexitropsin, an alkylator which exclusively induces 3-meA lesions. Indeed, MPG overexpression has been shown to increase the toxicity of alkylating agents that produce 7-meG adducts, and here we demonstrate that MPG-overexpressing cells have dramatically increased removal of 7-meG from their DNA. These data suggest that the mechanism of MPG-induced cytotoxicity involves the conversion of non-toxic 7-meG lesions into highly toxic repair intermediates. This study establishes a mechanism by which a benign DNA modification can be made toxic through the overexpression of an otherwise well-tolerated gene product, and the application of this principle could lead to improved chemotherapeutic strategies that reduce the peripheral toxicity of alkylating agents.


Subject(s)
Alkylating Agents/toxicity , DNA Adducts/metabolism , DNA Damage , DNA Glycosylases/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , Cell Line, Tumor , DNA Glycosylases/genetics , Gene Expression , Humans , Kinetics , Methyl Methanesulfonate/toxicity
7.
Br J Cancer ; 92(2): 334-41, 2005 Jan 31.
Article in English | MEDLINE | ID: mdl-15655552

ABSTRACT

Germline mutations have been associated with generation of various types of tumour. In this study, we investigated genetic alteration of germline tumours that affect the drug sensitivity of cells. Although all germline tumour cells we tested were hypersensitive to DNA-damaging drugs, no significant alteration was observed in their DNA repair activity or the expression of DNA repair proteins. In contrast, germline tumours expressed very low level of epidermal growth factor receptor (EGFR) compared to drug-resistant ovarian cancer cells. An immunohistochemical analysis indicated that most of the primary germline tumours we tested expressed very low level of EGFR. In accordance with this, overexpression of EGFR in germline tumour cells showed an increase in drug resistance, suggesting that a lack of EGFR, at least in part, contributes to the drug sensitivity of germline tumours.


Subject(s)
Drug Resistance, Neoplasm/physiology , ErbB Receptors/biosynthesis , Neoplasms, Germ Cell and Embryonal/genetics , Blotting, Western , DNA Repair/physiology , DNA Repair Enzymes/biosynthesis , Female , Humans , Immunohistochemistry , Neoplasms, Germ Cell and Embryonal/drug therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Tumor Cells, Cultured
8.
Am J Respir Cell Mol Biol ; 25(6): 692-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11726394

ABSTRACT

Bleomycin is a well-established anti-tumor drug. Its major untoward effect, pulmonary toxicity, has limited its usage. In this study, we used a DNA repair protein, yeast apurinic/apyrimidinic endonuclease (APN1) to reduce the toxicity of bleomycin on lung cells. A549 cells, an alveolar epithelial cell line, were transduced by MIEG3 retroviral vector encoding both enhanced green fluorescent protein (EGFP) and APN1. Transduced cells were sorted by fluorescent-activated cell sorter (FACS) analysis and were cloned. The APN1 expression of transduced A549 cell population and four selected clones expressing different levels of EGFP was confirmed by Northern, Western, and apurinic/apyrimidinic (AP) endonuclease activity analyses. The expression of APN1 was positively correlated with the expression of EGFP. The protective effect of APN1 against bleomycin was determined by single cell gel electrophoresis/Comet assay and by clonogenic survival assay following bleomycin treatment. The A549 population expressing APN1 showed a significant reduction of DNA damage in the presence of 20, 50, and 100 microg/ml bleomycin; similarly, the APN1-expressing A549 population also demonstrated increased survival in the presence of bleomycin compared with the vector-transduced A549 population. In selected clones, three of four APN1-expressing clones resulted in significantly improved cell survival. The current study suggests that the yeast DNA repair protein, APN1, can reduce bleomycin toxicity to target lung cells.


Subject(s)
Bleomycin/toxicity , DNA Damage , DNA Repair/physiology , Endodeoxyribonucleases/physiology , Mutagens/toxicity , Pulmonary Alveoli/drug effects , Saccharomyces cerevisiae Proteins/physiology , Bleomycin/pharmacology , Cell-Free System , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Clone Cells/drug effects , Clone Cells/metabolism , Colony-Forming Units Assay , Comet Assay , DNA/drug effects , DNA/metabolism , DNA Repair Enzymes , Dose-Response Relationship, Drug , Endodeoxyribonucleases/biosynthesis , Endodeoxyribonucleases/genetics , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins , Humans , Iron/metabolism , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mutagens/pharmacology , Oxidation-Reduction , Pulmonary Alveoli/cytology , Pulmonary Alveoli/enzymology , Recombinant Fusion Proteins , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Transfection
9.
Antioxid Redox Signal ; 3(4): 671-83, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11554453

ABSTRACT

The second enzyme in the DNA base excision repair (BER) pathway, apurinic/apyrimidinic (AP) endonuclease or Ape1, hydrolyzes the phosphodiester backbone immediately 5' to an AP site generating a normal 3'-hydroxyl group and an abasic deoxyribose-5-phosphate, which is processed by subsequent enzymes of the BER pathway. AP sites are the most common form of DNA damage, and the persistence of AP sites in DNA results in a block to DNA replication, cytotoxic mutations, and genetic instability. Interestingly, Ape1/ref-1 is a multifunctional protein that not only is a DNA repair enzyme, but also functions as a redox factor maintaining transcription factors, such as Fos, Jun, nuclear factor-kappaB, PAX (paired box-containing family of genes), hypoxia inducible factor-lalpha (HIF-1alpha), HIF-1-like factor, and p53, in an active reduced state. Apel/ref-1 has also been implicated in a number of other activities, one of which is the activation of bioreductive drugs requiring reduction for activity. In this report, we present data supporting our findings that another level of posttranslational modification of Apel/ref-1 that clearly affects the AP endonuclease activity is the reduction or oxidation of this protein. Furthermore, we show data demonstrating that at least one of the sites involved in this redox regulation is the cysteine amino acid found at position 310, immediately adjacent to the crucial histidine residue at position 309 in the DNA repair active site. These findings suggest that the Apel/ref-1 protein may be much more intimately regulated at the posttranslational level than initially imagined.


Subject(s)
Carbon-Oxygen Lyases/physiology , DNA Repair , Protein Processing, Post-Translational , Apurinic Acid/metabolism , Carbon-Oxygen Lyases/chemistry , Carbon-Oxygen Lyases/genetics , Cysteine/chemistry , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase , Diamide/pharmacology , Dithiothreitol/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Kinetics , Models, Biological , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/metabolism , Oxidants/pharmacology , Oxidation-Reduction , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Structure-Activity Relationship , Transcription Factors/metabolism
10.
Int J Oncol ; 19(4): 799-802, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562758

ABSTRACT

The multifunctional apurinic/apyrimidinic endonuclease (Ape1/ref-1) plays a key role in the human DNA base excision repair pathway. Ape1/ref-1 has also been shown to be involved in the redox control of transactivation activities of hypoxia-inducible factor (HIF)-1alpha. The aim of our study was to investigate the expression of these proteins in early stage invasive cervical cancer. Expression of Ape1/ref-1 and HIF-1alpha was detected immunohistochemically in 88 samples of cervical cancer stage pT1b. The levels of the proteins were compared and the prognostic influence of Ape1/ref-1 expression was investigated. Strong nuclear expression of Ape1/ref-1 was observed in 9 cases (10.2%), moderate in 22 cases (25%), weak in 17 cases (19.3%), and absent in 40 cases (45.5%). Furthermore, no correlation between Ape1/ref-1 and HIF-1alpha expression was observed (p=0.864). We also found no relationship of Ape1/ref-1 expression and survival (p>0.05, log-rank test). From these studies, we have concluded that in cervical cancer there is no correlation between the upstream redox regulatory protein of HIF-1, i.e., Ape1/ref-1, and HIF-1alpha expression. However, these studies do not address any functional relationship between the two proteins.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Nuclear Proteins/metabolism , Transcription Factors , Uterine Cervical Neoplasms/metabolism , Female , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunoenzyme Techniques , Neoplasm Staging , Oxidation-Reduction , Prognosis , Uterine Cervical Neoplasms/pathology
11.
Nucleic Acids Res ; 29(14): 3116-22, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11452037

ABSTRACT

Reactive oxygen species (ROS) arise through normal cellular aerobic respiration, and, in combination with external sources such as ionizing radiation, cigarette tar and smoke, and particulate matter generated by combustion, can have a profound negative effect on cellular macromolecules such as DNA that may lead to a number of human pathological disorders including accelerated aging and cancer. A major end product of ROS damage to DNA is the formation of apurinic/apyrimidinic (AP) sites, which without removal are known to halt mRNA and DNA synthesis, or act as non-coding lesions resulting in the increased generation of DNA mutations. In human cells, the major enzyme in correcting the deleterious effects of AP sites in DNA is through the participation of AP endonuclease (APE), which initiates the removal of baseless sites in DNA through the catalytic scission of the phosphodiester bond 5' and adjacent to an AP site. Interestingly, APE also possesses an activity (Ref-1) that controls the redox status of a number of transcription factors including Fos and Jun. The means by which APE/Ref-1 is directed to carry out such disparate roles are unknown. The presence of a number of phosphorylation sites scattered throughout both functional domains of APE/Ref-1 however offered one possible mechanism that we reasoned could play a role in dictating how this protein responds to different stimuli. Here we show that the in vitro redox activity of APE/Ref-1 is stimulated by PKC phosphorylation. Furthermore, when human cells were exposed to the PKC activator phorbol 12-myristate 13-acetate, an increase in redox activity was observed that corresponded to an increase in the phosphorylation status of APE/Ref-1. Importantly, human cells exposed to the oxidizing agent hypochlorite, followed by methyl methanesulfanate, responded with an increase in redox activity by APE/Ref-1 that also involved an increase in PKC activity and a corresponding increase in the phosphorylation of APE/Ref-1. These results suggest that the ability of APE/Ref-1 to perform its in vivo redox function is correlated to its susceptibility to PKC phosphorylation that notably occurs in response to DNA damaging agents.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , DNA Damage , Enzyme Activation , Humans , Hypochlorous Acid/pharmacology , K562 Cells , Methyl Methanesulfonate/pharmacology , Oxidation-Reduction , Phosphorylation/drug effects , Tetradecanoylphorbol Acetate/pharmacology
12.
Cancer Res ; 61(13): 5116-25, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431349

ABSTRACT

Modulation of DNA damage repair activity could lead to new approaches to reduce cytotoxic side effects of chemotherapy. N,N',N"-Triethylenethiophosphoramide (thioTEPA) induces the formation of amino-ethyl adducts of guanine, resulting in imidazole ring opening [formamidopyrimidine (Fapy)] and is associated with significant myelosuppression in dose-intensive therapies. In Escherichia coli, Fapy lesions are repaired by the Fapy-DNA glycosylase (Fpg) protein. We hypothesized that the expression of the Fpg could increase resistance of hematopoietic cells to thioTEPA-induced cytotoxicity. Expression of Fpg in bone marrow (BM) cells via a retrovirus vector was associated with demonstrable 8-oxodeoxyguanosine DNA glycosylase activity. BM cells were infected with a recombinant retrovirus, SF91, containing the Fpg gene and expressing the enhanced green fluorescence protein (EGFP) via an internal ribosomal entry site element. Control mice received BM transduced with the backbone containing IRES-EGFP alone. Fpg-transduced and GFP+ BM hematopoietic cells were resistant in vitro to thioTEPA at multiple concentrations. Mice transplanted with transduced cells were treated with four doses of thioTEPA (10 mg/kg) given over 7 weeks. Despite low transduction efficiency, peripheral blood leukocytes, hemoglobin, and platelet counts of thioTEPA-treated Fpg mice were significantly higher than treated control mice (P < 0.05). In addition, after treatment, the BM, spleen, and thymic cellularity as well as the number of GFP+ progenitor cells in the BM of treated mice were significantly higher than those of control group. Selection of Fpg-transduced cells in vivo was demonstrated by an increase in the mean fluorescence intensity of peripheral mononuclear cells of Fpg mice compared with pretreatment value. In addition, a significant increase in the EGFP-bright cells was demonstrated, suggesting preferential survival of high-expressing hematopoietic cells. Similar results were demonstrated in vitro with primary BM expressing the human functional counterpart of Fpg, OGG1. These results show that expression of the Fpg or hOGG1 protein protects hematopoietic cells from thioTEPA-induced DNA damage and suggest that a high level of expression of these repair proteins is required to establish resistance to this drug. Expression of Fpg and/or OGG1 may provide an novel approach to preventing thioTEPA-induced toxicity of primary hematopoietic cells.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , DNA Repair , Escherichia coli Proteins , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , N-Glycosyl Hydrolases/biosynthesis , N-Glycosyl Hydrolases/genetics , Thiotepa/toxicity , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , DNA-Formamidopyrimidine Glycosylase , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , N-Glycosyl Hydrolases/blood , Proviruses/genetics , Retroviridae/genetics , Spleen/cytology , Spleen/drug effects , Thymus Gland/cytology , Thymus Gland/drug effects , Transduction, Genetic , Virus Integration
13.
Nucleic Acids Res ; 29(12): 2558-66, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11410664

ABSTRACT

DNA repair status plays a major role in mutagenesis, carcinogenesis and resistance to genotoxic agents. Because DNA repair processes involve multiple enzymatic steps, understanding cellular DNA repair status has required several assay procedures. We have developed a novel in vitro assay that allows quantitative measurement of alkylation repair via O(6)-methylguanine DNA methyltransferase (MGMT) and base excision repair (BER) involving methylpurine DNA glycosylase (MPG), human 8-oxoguanine DNA glycosylase (hOGG1) and yeast and human abasic endonuclease (APN1 and APE/ref-1, respectively) from a single cell extract. This approach involves preparation of cell extracts in a common buffer in which all of the DNA repair proteins are active and the use of fluorometrically labeled oligonucleotide substrates containing DNA lesions specific to each repair protein. This method enables methylation and BER capacities to be determined rapidly from a small amount of starting sample. In addition, the stability of the fluorometric oligonucleotides precludes the substrate variability caused by continual radiolabeling. In this report this technique was applied to human breast carcinoma MDA-MB231 cells overexpressing human MPG in order to assess whether up-regulation of the initial step in BER alters the activity of selected other BER (hOGG1 and APE/ref-1) or direct reversal (MGMT) repair activities.


Subject(s)
Breast Neoplasms/enzymology , Carbon-Oxygen Lyases/metabolism , DNA Repair/genetics , Gene Expression Regulation, Enzymologic , N-Glycosyl Hydrolases/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylation , Breast Neoplasms/genetics , Buffers , Carbon-Oxygen Lyases/genetics , DNA Glycosylases , DNA Methylation , DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA-Formamidopyrimidine Glycosylase , Deoxyribonuclease IV (Phage T4-Induced) , Fluorescence , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , N-Glycosyl Hydrolases/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Oligonucleotides/genetics , Sensitivity and Specificity , Transfection , Tumor Cells, Cultured , Up-Regulation
14.
J Biol Chem ; 276(29): 27591-6, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11353770

ABSTRACT

The Drosophila S3 ribosomal protein has important roles in both protein translation and DNA repair. In regards to the latter activity, it has been shown that S3 contains vigorous N-glycosylase activity for the removal of 8-oxoguanine residues in DNA that leaves baseless sites in their places. Drosophila S3 also possesses an apurinic/apyrimidinic (AP) lyase activity in which the enzyme catalyzes a beta-elimination reaction that cleaves phosphodiester bonds 3' and adjacent to an AP lesion in DNA. In certain situations, this is followed by a delta-elimination reaction that ultimately leads to the formation of a single nucleotide gap in DNA bordered by 5'- and 3'-phosphate groups. The human S3 protein, although 80% identical to its Drosophila homolog and shorter by only two amino acids, has only marginal N-glycosylase activity. Its lyase activity only cleaves AP DNA by a beta-elimination reaction, thus further distinguishing itself from the Drosophila S3 protein in lacking a delta-elimination activity. Using a hidden Markov model analysis based on the crystal structures of several DNA repair proteins, the enzymatic differences between Drosophila and human S3 were suggested by the absence of a conserved glutamine residue in human S3 that usually resides at the cleft of the deduced active site pocket of DNA glycosylases. Here we show that the replacement of the Drosophila glutamine by an alanine residue leads to the complete loss of glycosylase activity. Unexpectedly, the delta-elimination reaction at AP sites was also abrogated by a change in the Drosophila glutamine residue. Thus, a single amino acid change converted the Drosophila activity into one that is similar to that possessed by the human S3 protein. In support of this were experiments executed in vivo that showed that human S3 and the Drosophila site-directed glutamine-changed S3 performed poorly when compared with Drosophila wild-type S3 and its ability to protect a bacterial mutant from the harmful effects of DNA-damaging agents.


Subject(s)
DNA Repair , Guanine/analogs & derivatives , Guanine/metabolism , Ribosomal Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Borohydrides/chemistry , Catalysis , DNA/metabolism , DNA Damage , DNA Primers , Drosophila , Humans , Mutagenesis, Site-Directed , Mutagens/toxicity , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Sequence Homology, Amino Acid
15.
Clin Cancer Res ; 7(4): 824-30, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309329

ABSTRACT

The DNA base excision repair pathway is responsible for the repair of cellular alkylation and oxidative DNA damage. A crucial step in the BER pathway involves the cleavage of baseless sites in DNA by an apurinic/apyrimidinic or baseless (AP) endonuclease (Ape1/ref-1), which is a multifunctional enzyme that acts not only as an AP endonuclease but also as a redox-modifying factor for a variety of transcription factors including Fos, Jun, paired box containing genes (PAX), nuclear factor-kappaB, hypoxia-inducible factor alpha (HIF-1alpha), HIF-like factor (HLF), p53, and others. The expression of Ape1/ref-1 in prostate has not been characterized previously. Ape1/ref-1 nuclear immunohistochemistry levels, scored for intensity as 1+, 2+, or 3+, were 91, 3, and 6% in benign hypertrophy (BPH), 0, 42, and 58% in prostatic intraepithelial neoplasia (PIN) and 3, 30, and 67% in prostate cancer, respectively, clearly showing an increase in Ape1/ref-1 nuclear staining in the PIN and cancer compared with BPH. Furthermore, the level of cytoplasmic staining of Ape1/ref-1 in cancer and PIN were elevated (42 and 36%, respectively) compared with BPH (5%). There was no correlation with prostate-specific antigen values or doubling times to Ape1/ref-1 levels. In conclusion, we have demonstrated that Ape1/ref-1 is dramatically elevated in prostate cancer, the level of staining of Ape1/ref-1 increases from low in BPH to intense in PIN and cancer, and there is an increase in the amount of Ape1/ref-1 in the cytoplasm of PIN and cancer compared with BPH. Given these results, we conclude that Ape1/ref-1 may be a diagnostic marker for early prostate cancer and play a role, through its repair, redox, or both functions, in the physiology of the early development of prostate cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Carbon-Oxygen Lyases/metabolism , DNA Ligases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Prostatic Neoplasms/enzymology , Humans , Male , Oncogene Proteins/metabolism , Oxidation-Reduction , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
16.
Am J Physiol Lung Cell Mol Physiol ; 280(4): L755-61, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238017

ABSTRACT

1,3-Bis(2-chloroethyl)-1-nitrosourea (BCNU) is an important cause of pulmonary toxicity. BCNU alkylates DNA at the O(6) position of guanine. O(6)-methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein that removes alkyl groups from the O(6) position of guanine. To determine whether overexpression of MGMT in a lung cell reduces BCNU toxicity, the MGMT gene was transfected into A549 cells, a lung epithelial cell line. Transfected A549 cell populations demonstrated high levels of MGMT RNA, MGMT protein, and DNA repair activity. The overexpression of MGMT in lung epithelial cells provided protection from the cytotoxic effects of BCNU. Control A549 cells incubated with 100 microM BCNU had a cell survival rate of 12.5 +/- 1.2%; however, A549 cells overexpressing MGMT had a survival rate of 71.8 +/- 2.7% (P < 0.001). We also demonstrated successful transfection of MGMT into human pulmonary artery endothelial cells and a primary culture of rat type II alveolar epithelial cells with overexpression of MGMT, resulting in significant protection from BCNU toxicity. These data suggest that overexpression of DNA repair proteins such as MGMT in lung cells may protect the lung cells from cytotoxic effects of cancer chemotherapy drugs such as BCNU.


Subject(s)
Carmustine/poisoning , Lung/drug effects , O(6)-Methylguanine-DNA Methyltransferase/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , DNA Repair , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Lung/cytology , O(6)-Methylguanine-DNA Methyltransferase/genetics , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiology , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , RNA, Messenger/metabolism , Rats , Transfection
17.
Cancer Res ; 61(5): 2220-5, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280790

ABSTRACT

The human AP endonuclease (Ape1 or ref-1) DNA base excision repair (BER) enzyme is a multifunctional protein that has an impact on a wide variety of important cellular functions including oxidative signaling, transcription factor regulation, and cell cycle control. It acts on mutagenic AP (baseless) sites in DNA as a critical member of the DNA BER repair pathway. Moreover, Ape1/ref-1 stimulates the DNA-binding activity of transcription factors (Fos-Jun, nuclear factor-kappaB, Myb, ATF/cyclic AMP-responsive element binding protein family, HIF-1alpha, HLF, PAX, and p53) through a redox mechanism and thus represents a novel component of signal transduction processes that regulate eukaryotic gene expression. Ape1/ref-1 has also been shown to be closely linked to apoptosis associated with thioredoxin, and altered levels of Ape1/ref-1 have been found in some cancers. In a pilot study, we have examined Ape1/ref-1 expression by immunohistochemistry in sections of germ cell tumors (GCTs) from 10 patients with testicular cancer of various histologies including seminomas, yolk sac tumors, and malignant teratomas. Ape1/ref-1 was expressed at relatively high levels in the tumor cells of nearly all sections. We hypothesized that elevated expression of Ape1/ref-1 is responsible in part for the resistance to therapeutic agents. To answer this hypothesis, we overexpressed the Ape1/ref-1 cDNA in the GCT cell line NT2/D1 using retroviral gene transduction with the vector LAPESN. Using an oligonucleotide cleavage assay and immunohistochemistry to assess Ape1/ref-1 repair activity and expression, respectively, we found that the repair activity and relative Ape1/ref-1 expression in GCT cell lines are directly related. NT2/D1 cells transduced with Ape1/ref-1 exhibited 2-fold higher AP endonuclease activity in the oligonucleotide cleavage assay, and this was reflected in a 2-3-fold increase in protection against bleomycin. Lesser protection was observed with gamma-irradiation. We conclude that: (a) Ape1/ref-1 is expressed at relatively high levels in some GCTs; (b) elevated expression of Ape1/ref-1 in testicular cancer cell lines results in resistance to certain therapeutic agents; and (c) Ape1/ref-1 expression in GCT cell lines determined by immunohistochemistry and repair activity assays parallels the level of protection from bleomycin. We further hypothesize that elevated Ape1/ref-1 levels observed in human testicular cancer may be related to their relative resistance to therapy and may serve as a diagnostic marker for refractory disease. To our knowledge, this is the first example of overexpressing Ape1/ref-1 in a mammalian system resulting in enhanced protection to DNA-damaging agents.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Bleomycin/pharmacology , Carbon-Oxygen Lyases/biosynthesis , Carcinoma, Embryonal/metabolism , Germinoma/metabolism , Radiation Tolerance/physiology , Carbon-Oxygen Lyases/genetics , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/radiotherapy , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease IV (Phage T4-Induced) , Drug Resistance, Neoplasm , Gene Transfer Techniques , Germinoma/drug therapy , Germinoma/radiotherapy , Humans , Retroviridae/genetics , Tumor Cells, Cultured
18.
J Pharmacol Exp Ther ; 296(3): 825-31, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181913

ABSTRACT

Chemotherapeutic agents used in the treatment of cancer often lead to dose-limiting bone marrow suppression and may initiate secondary leukemia. N,N',N"-triethylenethiophosphoramide (thiotepa), a polyfunctional alkylating agent, is used in the treatment of breast, ovarian, and bladder carcinomas and is also being tested for efficacy in the treatment of central nervous system tumors. Thiotepa produces ring-opened bases such as formamidopyrimidine and 7-methyl-formamidopyrimidine, which can be recognized and repaired by the formamidopyrimidine glycosylase/AP lyase (Fpg) enzyme of Escherichia coli. Using this background information, we have created constructs using the E. coli fpg gene along with the functional equivalent human ortholog alpha-hOgg1. Although protection with the Fpg protein has been previously observed in Chinese hamster ovary cells, we demonstrate significant (100-fold) protection against thiotepa using the E. coli Fpg or the human alpha-hOgg1 cDNA in NIH3T3 cells. We have also observed a 10-fold protection by both the Fpg and alpha-hOgg1 transgenes against 1,3-N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU) and, to a lesser extent, mafosfamide (2-fold), an active form of the clinical agent cyclophosphamide. These latter two findings are novel and are particularly significant since the added protection was in an O(6)-methylguanine-DNA methyltransferase-positive background. These results support our general approach of using DNA base excision repair genes in gene therapy for cellular protection of normal cells during chemotherapy, particularly against the severe myelosuppressive effect of agents such as thiotepa, BCNU, and cyclophosphamide.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/pharmacology , DNA Repair , DNA/drug effects , Escherichia coli Proteins , Genetic Therapy , N-Glycosyl Hydrolases/pharmacology , Thiotepa/pharmacology , 3T3 Cells , Animals , Antineoplastic Agents/toxicity , Blotting, Northern , Carmustine/pharmacology , Cell Division/drug effects , Cyclophosphamide/toxicity , Cytoprotection , DNA/metabolism , DNA Damage , DNA-Formamidopyrimidine Glycosylase , Drug Interactions , Genetic Vectors , Humans , Mice , N-Glycosyl Hydrolases/genetics , Transfection , Transgenes
19.
Mutat Res ; 485(2): 107-19, 2001 Mar 07.
Article in English | MEDLINE | ID: mdl-11182542

ABSTRACT

Cells harvested from Fanconi anemia (FA) patients show an increased hypersensitivity to the multifunctional DNA damaging agent mitomycin C (MMC), which causes cross-links in DNA as well as 7,8-dihydro-8-oxoguanine (8-oxoG) adducts indicative of escalated oxidative DNA damage. We show here that the Drosophila multifunctional S3 cDNA, which encodes an N-glycosylase/apurinic/apyrimidinic (AP) lyase activity was found to correct the FA Group A (FA(A)) and FA Group C (FA(C)) sensitivity to MMC and hydrogen peroxide (H2O2). Furthermore, the Drosophila S3 cDNA was shown to protect AP endonuclease deficient E. coli cells against H(2)O(2) and MMC, and also protect 8-oxoG repair deficient mutM E. coli strains against MMC and H2O2 cell toxicity. Conversely, the human S3 protein failed to complement the AP endonuclease deficient E. coli strain, most likely because it lacks N-glycosylase activity for the repair of oxidatively-damaged DNA bases. Although the human S3 gene is clearly not the genetic alteration in FA cells, our results suggest that oxidative DNA damage is intimately involved in the overall FA phenotype, and the cytotoxic effect of selective DNA damaging agents in FA cells can be overcome by trans-complementation with specific DNA repair cDNAs. Based on these findings, we would predict other oxidative repair proteins, or oxidative scavengers, could serve as protective agents against the oxidative DNA damage that occurs in FA.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA, Complementary/pharmacology , Escherichia coli Proteins , Fanconi Anemia/metabolism , Guanine/analogs & derivatives , Ribosomal Proteins/metabolism , Animals , Antigens, CD34/metabolism , Carbon-Oxygen Lyases/deficiency , Carbon-Oxygen Lyases/genetics , Cell Survival/drug effects , Cells, Cultured , Cross-Linking Reagents/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/drug effects , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA-Formamidopyrimidine Glycosylase , Deoxyribonuclease IV (Phage T4-Induced) , Drosophila , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Fanconi Anemia/genetics , Gene Transfer Techniques , Genetic Complementation Test , Guanine/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mitomycin/pharmacology , N-Glycosyl Hydrolases/deficiency , N-Glycosyl Hydrolases/metabolism , Oxidation-Reduction/drug effects , Retroviridae/genetics , Ribosomal Proteins/pharmacology
20.
Mutat Res ; 485(2): 143-52, 2001 Mar 07.
Article in English | MEDLINE | ID: mdl-11182545

ABSTRACT

Mutations of mitochondrial DNA (mtDNA) are associated with different human diseases, including cancer and aging. Reactive oxygen species produced during oxidative phosphorylation are a major source of mtDNA damage. It is not clear, however, whether DNA repair mechanisms, able to abolish effects due to oxidative damage, are present in mitochondria. APE/Ref-1 is a nuclear protein possessing both redox activity (by which activates, "in vitro", the DNA-binding functions of several transcription factors) and DNA repair activity over apurinic/apyrimidinic sites. Immunohistochemical evidences indicate that in follicular thyroid cells, APE/Ref-1 is located in both nucleus and cytoplasm. Electronmicroscopy immunocytochemistry performed in the rat thyroid FRTL-5 cell line, indicates that part of the cytoplasmatic APE/Ref-1 is located in mitochondria. The presence of APE/Ref-1 inside mitochondria is further demonstrated by western blot analysis after cell fractionation. In the Kimol cell line (which is derived from FRTL-5, transformed by the Ki-ras oncogene) the amount of mitochondrial APE/Ref-1 is reduced by three to fourfold with respect to the normal FRTL-5 cells. These results suggest that: (i) a machinery capable of repairing DNA damaged by oxidative stress is present in mitochondria and (ii) mtDNA repair mechanisms may be impaired during cell transformation.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Mitochondria/metabolism , Thyroid Gland/metabolism , Animals , Carbon-Oxygen Lyases/analysis , Cell Line , DNA Repair , DNA, Mitochondrial/metabolism , Immunohistochemistry , Microscopy, Electron , Microscopy, Fluorescence , Mitochondria/chemistry , Mitochondria/ultrastructure , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism , Subcellular Fractions/metabolism , Thyroid Gland/cytology , Thyroid Gland/ultrastructure , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL