Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
J Toxicol Sci ; 46(6): 283-288, 2021.
Article in English | MEDLINE | ID: mdl-34078835

ABSTRACT

Abasic sites are formed in cells by various factors including environmental mutagens and considered to be involved in cancer initiation, promotion, and progression. A chemically stable abasic site analog (tetrahydrofuran-type analog, THF) induces untargeted base substitutions as well as targeted substitution and large deletion mutations in human cells. The untargeted substitutions may be initiated by the cleavage of the DNA strand bearing THF by the human apurinic/apyrimidinic endonuclease 1 (APE1) protein, the major repair enzyme for THF and abasic sites. To examine the effects of lower APE1 levels, the protein was knocked down by siRNA in human U2OS cells. A plasmid containing a single THF modification outside the supF gene was introduced into the knockdown cells, and the untargeted substitution mutations in the reporter gene were analyzed. Unexpectedly, the knockdown had no evident impact on their frequency and spectrum. The G bases of 5'-GpA-3' dinucleotides on the modified strand were quite frequently substituted, with and without the APE1 knockdown. These results suggested that the DNA strand cleavage by APE1 is not essential for the THF-induced untargeted base substitutions.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , DNA/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Furans/metabolism , Gene Knockdown Techniques , Genes, Reporter/genetics , Humans , Mutation , Plasmids/metabolism
2.
Mutagenesis ; 35(1): 129-149, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31858150

ABSTRACT

Alterations of DNA repair enzymes and consequential triggering of aberrant DNA damage response (DDR) pathways are thought to play a pivotal role in genomic instabilities associated with cancer development, and are further thought to be important predictive biomarkers for therapy using the synthetic lethality paradigm. However, novel unpredicted perspectives are emerging from the identification of several non-canonical roles of DNA repair enzymes, particularly in gene expression regulation, by different molecular mechanisms, such as (i) non-coding RNA regulation of tumour suppressors, (ii) epigenetic and transcriptional regulation of genes involved in genotoxic responses and (iii) paracrine effects of secreted DNA repair enzymes triggering the cell senescence phenotype. The base excision repair (BER) pathway, canonically involved in the repair of non-distorting DNA lesions generated by oxidative stress, ionising radiation, alkylation damage and spontaneous or enzymatic deamination of nucleotide bases, represents a paradigm for the multifaceted roles of complex DDR in human cells. This review will focus on what is known about the canonical and non-canonical functions of BER enzymes related to cancer development, highlighting novel opportunities to understand the biology of cancer and representing future perspectives for designing new anticancer strategies. We will specifically focus on APE1 as an example of a pleiotropic and multifunctional BER protein.


Subject(s)
DNA Repair Enzymes/physiology , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Neoplasms/enzymology , DNA/metabolism , DNA Damage , DNA Repair Enzymes/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
3.
Biochemistry ; 58(35): 3646-3655, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31407575

ABSTRACT

Deciphering factors modulating DNA repair in chromatin is of great interest because nucleosomal positioning influences mutation rates. H3K56 acetylation (Ac) is implicated in chromatin landscape regulation, impacting genomic stability, yet the effect of H3K56Ac on DNA base excision repair (BER) remains unclear. We determined whether H3K56Ac plays a role in regulating AP site incision by AP endonuclease 1 (APE1), an early step in BER. Our in vitro studies of acetylated, well-positioned nucleosome core particles (H3K56Ac-601-NCPs) demonstrate APE1 strand incision is enhanced compared with that of unacetylated WT-601-NCPs. The high-mobility group box 1 protein enhances APE1 activity in WT-601-NCPs, but this effect is not observed in H3K56Ac-601-NCPs. Therefore, our results suggest APE1 activity on NCPs can be modulated by H3K56Ac.


Subject(s)
DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Histone Acetyltransferases/metabolism , Histones/metabolism , Nucleosomes/metabolism , Acetylation , Animals , Binding Sites/genetics , DNA Repair/genetics , Escherichia coli , Genomic Instability , Histones/chemistry , Humans , Lysine/metabolism , Methanosarcina barkeri , Mice , Nucleosomes/genetics , Protein Binding , Protein Processing, Post-Translational/physiology , Sirtuins/genetics , Sirtuins/metabolism , Xenopus laevis
4.
Nat Struct Mol Biol ; 25(10): 971-980, 2018 10.
Article in English | MEDLINE | ID: mdl-30291363

ABSTRACT

The Ku70-Ku80 (Ku) heterodimer binds rapidly and tightly to the ends of DNA double-strand breaks and recruits factors of the non-homologous end-joining (NHEJ) repair pathway through molecular interactions that remain unclear. We have determined crystal structures of the Ku-binding motifs (KBM) of the NHEJ proteins APLF (A-KBM) and XLF (X-KBM) bound to a Ku-DNA complex. The two KBM motifs bind remote sites of the Ku80 α/ß domain. The X-KBM occupies an internal pocket formed by an unprecedented large outward rotation of the Ku80 α/ß domain. We observe independent recruitment of the APLF-interacting protein XRCC4 and of XLF to laser-irradiated sites via binding of A- and X-KBMs, respectively, to Ku80. Finally, we show that mutation of the X-KBM and A-KBM binding sites in Ku80 compromises both the efficiency and accuracy of end joining and cellular radiosensitivity. A- and X-KBMs may represent two initial anchor points to build the intricate interaction network required for NHEJ.


Subject(s)
DNA End-Joining Repair , DNA Repair Enzymes/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-Binding Proteins/chemistry , Ku Autoantigen/chemistry , Poly-ADP-Ribose Binding Proteins/chemistry , Conserved Sequence , Crystallography, X-Ray , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Humans , Ku Autoantigen/metabolism , Ku Autoantigen/physiology , Models, Molecular , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/physiology , Protein Domains
5.
J Invest Dermatol ; 137(2): 466-474, 2017 02.
Article in English | MEDLINE | ID: mdl-27729279

ABSTRACT

The phototherapeutic effects of visible red light on skin have been extensively investigated, but the underlying biological mechanisms remain poorly understood. We aimed to elucidate the protective mechanism of visible red light in terms of DNA repair of UV-induced oxidative damage in normal human dermal fibroblasts. The protective effect of visible red light on UV-induced DNA damage was identified by several assays in both two-dimensional and three-dimensional cell culture systems. With regard to the protective mechanism of visible red light, our data showed alterations in base excision repair mediated by growth arrest and DNA damage inducible, alpha (GADD45A). We also observed an enhancement of the physical activity of GADD45A and apurinic/apyrimidinic endonuclease 1 (APE1) by visible red light. Moreover, UV-induced DNA damages were diminished by visible red light in an APE1-dependent manner. On the basis of the decrease in GADD45A-APE1 interaction in the activating transcription factor-2 (ATF2)-knockdown system, we suggest a role for ATF2 modulation in GADD45A-mediated DNA repair upon visible red light exposure. Thus, the enhancement of GADD45A-mediated base excision repair modulated by ATF2 might be a potential protective mechanism of visible red light.


Subject(s)
Cell Cycle Proteins/physiology , Cytoprotection , DNA Repair , Light , Nuclear Proteins/physiology , Skin/radiation effects , Activating Transcription Factor 2/physiology , Cells, Cultured , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Fibroblasts/radiation effects , Humans , Skin/metabolism , Tumor Suppressor Protein p53/physiology
6.
Oncotarget ; 7(34): 54430-54444, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27303920

ABSTRACT

Ionizing radiation (IR) induces highly cytotoxic double-strand breaks (DSBs) and also clustered oxidized bases in mammalian genomes. Base excision repair (BER) of bi-stranded oxidized bases could generate additional DSBs as repair intermediates in the vicinity of direct DSBs, leading to loss of DNA fragments. This could be avoided if DSB repair via DNA-PK-mediated nonhomologous end joining (NHEJ) precedes BER initiated by NEIL1 and other DNA glycosylases (DGs). Here we show that DNA-PK subunit Ku inhibits DGs via direct interaction. The scaffold attachment factor (SAF)-A, (also called hnRNP-U), phosphorylated at Ser59 by DNA-PK early after IR treatment, is linked to transient release of chromatin-bound NEIL1, thus preventing BER. SAF-A is subsequently dephosphorylated. Ku inhibition of DGs in vitro is relieved by unphosphorylated SAF-A, but not by the phosphomimetic Asp59 mutant. We thus propose that SAF-A, in concert with Ku, temporally regulates base damage repair in irradiated cell genome.


Subject(s)
DNA Repair , Heterogeneous-Nuclear Ribonucleoprotein U/physiology , Ku Autoantigen/physiology , Radiation Injuries/etiology , DNA Breaks, Double-Stranded , DNA Glycosylases/physiology , DNA Repair Enzymes/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/physiology , HEK293 Cells , Humans , Phosphorylation , Radiation Tolerance
7.
Proc Natl Acad Sci U S A ; 113(25): E3558-67, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27274063

ABSTRACT

A major hallmark of oxidative DNA damage after stroke is the induction of apurinic/apyrimidinic (AP) sites and strand breaks. To mitigate cell loss after oxidative DNA damage, ischemic cells rapidly engage the base excision-repair proteins, such as the AP site-repairing enzyme AP endonuclease-1 (APE1), also named redox effector factor-1 (Ref-1). Although forced overexpression of APE1 is known to protect against oxidative stress-induced neurodegeneration, there is no concrete evidence demonstrating a role for endogenous APE1 in the long-term recovery of gray and white matter following ischemic injury. To address this gap, we generated, to our knowledge, the first APE1 conditional knockout (cKO) mouse line under control of tamoxifen-dependent Cre recombinase. Using a well-established model of transient focal cerebral ischemia (tFCI), we show that induced deletion of APE1 dramatically enlarged infarct volume and impaired the recovery of sensorimotor and cognitive deficits. APE1 cKO markedly increased postischemic neuronal and oligodendrocyte degeneration, demonstrating that endogenous APE1 preserves both gray and white matter after tFCI. Because white matter repair is instrumental in behavioral recovery after stroke, we also examined the impact of APE1 cKO on demyelination and axonal conduction and discovered that APE1 cKO aggravated myelin loss and impaired neuronal communication following tFCI. Furthermore, APE1 cKO increased AP sites and activated the prodeath signaling proteins, PUMA and PARP1, after tFCI in topographically distinct manners. Our findings provide evidence that endogenous APE1 protects against ischemic infarction in both gray and white matter and facilitates the functional recovery of the central nervous system after mild stroke injury.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Gray Matter/physiopathology , Stroke/physiopathology , White Matter/physiopathology , Animals , Behavior, Animal , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
PLoS One ; 10(11): e0143026, 2015.
Article in English | MEDLINE | ID: mdl-26565960

ABSTRACT

The Adaptor Protein (AP)-3 complex is an evolutionary conserved, molecular sorting device that mediates the intracellular trafficking of proteins to lysosomes and related organelles. Genetic defects in AP-3 subunits lead to impaired biogenesis of lysosome-related organelles (LROs) such as mammalian melanosomes and insect eye pigment granules. In this work, we have performed a forward screening for genetic modifiers of AP-3 function in the fruit fly, Drosophila melanogaster. Specifically, we have tested collections of large multi-gene deletions--which together covered most of the autosomal chromosomes-to identify chromosomal regions that, when deleted in single copy, enhanced or ameliorated the eye pigmentation phenotype of two independent AP-3 subunit mutants. Fine-mapping led us to define two non-overlapping, relatively small critical regions within fly chromosome 3. The first critical region included the Atg2 gene, which encodes a conserved protein involved in autophagy. Loss of one functional copy of Atg2 ameliorated the pigmentation defects of mutants in AP-3 subunits as well as in two other genes previously implicated in LRO biogenesis, namely Blos1 and lightoid, and even increased the eye pigment content of wild-type flies. The second critical region included the ArfGAP1 gene, which encodes a conserved GTPase-activating protein with specificity towards GTPases of the Arf family. Loss of a single functional copy of the ArfGAP1 gene ameliorated the pigmentation phenotype of AP-3 mutants but did not to modify the eye pigmentation of wild-type flies or mutants in Blos1 or lightoid. Strikingly, loss of the second functional copy of the gene did not modify the phenotype of AP-3 mutants any further but elicited early lethality in males and abnormal eye morphology when combined with mutations in Blos1 and lightoid, respectively. These results provide genetic evidence for new functional links connecting the machinery for biogenesis of LROs with molecules implicated in autophagy and small GTPase regulation.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , GTPase-Activating Proteins/physiology , Pigmentation/genetics , Animals , Autophagy , Autophagy-Related Proteins , Chromosome Mapping , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Drosophila melanogaster/physiology , Evolution, Molecular , Eye Proteins/genetics , Eye Proteins/physiology , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/physiology , GTPase-Activating Proteins/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Hemizygote , Lysosomes/metabolism , Male , Models, Genetic , Mutation , Phenotype , Photoreceptor Cells, Invertebrate/physiology , rab GTP-Binding Proteins
9.
Comb Chem High Throughput Screen ; 18(9): 881-91, 2015.
Article in English | MEDLINE | ID: mdl-26111950

ABSTRACT

Survival of cells and maintenance of genome depend on detection and repair of damaged DNA through intricate mechanisms. Cancer treatment relies on chemotherapy or radiation therapy that kills neoplastic cells by causing immense damage to the DNA. In many cases, escalated DNA repair mechanism leads to resistance against these therapies and therefore, there is a need to expand the interest in developing drugs that can sensitize the cells to such therapies by interfering with the DNA repair mechanism. Several studies have suggested a link between over expression of the primary mammalian enzyme, Apurinic/Apyrimidinic Endonuclease (APE1), responsible for abasic (or AP) site removal in the DNA and resistance of these cells to cancer therapy, whereas APE1 down-regulation sensitizes the cells to DNA damaging agents. Thus, the current treatment efficacy can be improved by aiding to selective sensitization of cancer cells and protection of normal cells. In the present study, we have used machine learning based approach by selecting assorted compounds with known activity for APE1 and constructed a range of in silico predictive classification models to discriminate between the inhibitors and non-inhibitors. These models can be applied to numerous other unscreened compounds to select the ones which are more likely to be the inhibitors for APE1. We have further found the common molecular substructures which were associated with the molecular activity of the compounds using a substructure search approach.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Informatics , Antineoplastic Agents/pharmacology , DNA Damage/drug effects , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Endonucleases , Multifunctional Enzymes
10.
J Biol Chem ; 289(44): 30635-30644, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25210033

ABSTRACT

High linear energy transfer (LET) radiation from space heavy charged particles or a heavier ion radiotherapy machine kills more cells than low LET radiation, mainly because high LET radiation-induced DNA damage is more difficult to repair. Relative biological effectiveness (RBE) is the ratio of the effects generated by high LET radiation to low LET radiation. Previously, our group and others demonstrated that the cell-killing RBE is involved in the interference of high LET radiation with non-homologous end joining but not homologous recombination repair. This effect is attributable, in part, to the small DNA fragments (≤40 bp) directly produced by high LET radiation, the size of which prevents Ku protein from efficiently binding to the two ends of one fragment at the same time, thereby reducing non-homologous end joining efficiency. Here we demonstrate that Ape1, an enzyme required for processing apurinic/apyrimidinic (known as abasic) sites, is also involved in the generation of small DNA fragments during the repair of high LET radiation-induced base damage, which contributes to the higher RBE of high LET radiation-induced cell killing. This discovery opens a new direction to develop approaches for either protecting astronauts from exposure to space radiation or benefiting cancer patients by sensitizing tumor cells to high LET radiotherapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Animals , Cell Death , Cell Line , DNA Fragmentation , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Humans , Linear Energy Transfer , MRE11 Homologue Protein , Mice, Inbred C57BL , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/metabolism , Up-Regulation , X-Rays
11.
Methods Mol Biol ; 1105: 159-68, 2014.
Article in English | MEDLINE | ID: mdl-24623227

ABSTRACT

Abasic sites in DNA arise under a variety of circumstances, including destabilization of bases through oxidative stress, as an intermediate in base excision repair, and through spontaneous loss. Their persistence can yield a blockade to RNA transcription and DNA synthesis and can be a source of mutations. Organisms have developed an enzymatic means of repairing abasic sites in DNA that generally involves a DNA repair pathway that is initiated by a repair protein creating a phosphodiester break ("nick") adjacent to the site of base loss. Here we describe a method for analyzing the manner in which repair endonucleases differ in the way they create nicks in DNA and how to distinguish between them using cellular crude extracts.


Subject(s)
DNA Cleavage , Animals , Apurinic Acid/genetics , DNA Damage , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Humans , Hydrolysis , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oxidative Stress
12.
PLoS One ; 9(3): e90261, 2014.
Article in English | MEDLINE | ID: mdl-24595271

ABSTRACT

Secondhand smoke (SHS) is a confirmed lung carcinogen that introduces thousands of toxic chemicals into the lungs. SHS contains chemicals that have been implicated in causing oxidative DNA damage in the airway epithelium. Although DNA repair is considered a key defensive mechanism against various environmental attacks, such as cigarette smoking, the associations of individual repair enzymes with susceptibility to lung cancer are largely unknown. This study investigated the role of NEIL2, a DNA glycosylase excising oxidative base lesions, in human lung cells treated with sidestream smoke (SSS), the main component of SHS. To do so, we generated NEIL2 knockdown cells using siRNA-technology and exposed them to SSS-laden medium. Representative SSS chemical compounds in the medium were analyzed by mass spectrometry. An increased production of reactive oxygen species (ROS) in SSS-exposed cells was detected through the fluorescent detection and the induction of HIF-1α. The long amplicon-quantitative PCR (LA-QPCR) assay detected significant dose-dependent increases of oxidative DNA damage in the HPRT gene of cultured human pulmonary fibroblasts (hPF) and BEAS-2B epithelial cells exposed to SSS for 24 h. These data suggest that SSS exposure increased oxidative stress, which could contribute to SSS-mediated toxicity. siRNA knockdown of NEIL2 in hPF and HEK 293 cells exposed to SSS for 24 h resulted in significantly more oxidative DNA damage in HPRT and POLB than in cells with control siRNA. Taken together, our data strongly suggest that decreased repair of oxidative DNA base lesions due to an impaired NEIL2 expression in non-smokers exposed to SSS would lead to accumulation of mutations in genomic DNA of lung cells over time, thus contributing to the onset of SSS-induced lung cancer.


Subject(s)
DNA Damage , DNA Glycosylases/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Oxidative Stress , Smoke/adverse effects , Base Sequence , Cell Line , Culture Media , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/cytology , Lung/metabolism , Lung Neoplasms/metabolism , RNA, Small Interfering , Reactive Oxygen Species/metabolism
13.
Antioxid Redox Signal ; 20(4): 678-707, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23834463

ABSTRACT

SIGNIFICANCE: Human apurinic/apyrimidinic endonuclease 1 (APE1, also known as REF-1) was isolated based on its ability to cleave at AP sites in DNA or activate the DNA binding activity of certain transcription factors. We review herein topics related to this multi-functional DNA repair and stress-response protein. RECENT ADVANCES: APE1 displays homology to Escherichia coli exonuclease III and is a member of the divalent metal-dependent α/ß fold-containing phosphoesterase superfamily of enzymes. APE1 has acquired distinct active site and loop elements that dictate substrate selectivity, and a unique N-terminus which at minimum imparts nuclear targeting and interaction specificity. Additional activities ascribed to APE1 include 3'-5' exonuclease, 3'-repair diesterase, nucleotide incision repair, damaged or site-specific RNA cleavage, and multiple transcription regulatory roles. CRITICAL ISSUES: APE1 is essential for mouse embryogenesis and contributes to cell viability in a genetic background-dependent manner. Haploinsufficient APE1(+/-) mice exhibit reduced survival, increased cancer formation, and cellular/tissue hyper-sensitivity to oxidative stress, supporting the notion that impaired APE1 function associates with disease susceptibility. Although abnormal APE1 expression/localization has been seen in cancer and neuropathologies, and impaired-function variants have been described, a causal link between an APE1 defect and human disease remains elusive. FUTURE DIRECTIONS: Ongoing efforts aim at delineating the biological role(s) of the different APE1 activities, as well as the regulatory mechanisms for its intra-cellular distribution and participation in diverse molecular pathways. The determination of whether APE1 defects contribute to human disease, particularly pathologies that involve oxidative stress, and whether APE1 small-molecule regulators have clinical utility, is central to future investigations.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Animals , Antineoplastic Agents/pharmacology , Catalytic Domain , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Humans , Molecular Targeted Therapy , Mutation, Missense , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Protein Structure, Secondary , RNA Cleavage
14.
Antioxid Redox Signal ; 20(4): 708-26, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23901781

ABSTRACT

SIGNIFICANCE: Reactive oxygen species (ROS) are generated by exogenous and environmental genotoxins, but also arise from mitochondria as byproducts of respiration in the body. ROS generate DNA damage of which pathological consequence, including cancer is well established. Research efforts are intense to understand the mechanism of DNA base excision repair, the primary mechanism to protect cells from genotoxicity caused by ROS. RECENT ADVANCES: In addition to the notion that oxidative DNA damage causes transformation of cells, recent studies have revealed how the mitochondrial deficiencies and ROS generation alter cell growth during the cancer transformation. CRITICAL ISSUES: The emphasis of this review is to highlight the importance of the cellular response to oxidative DNA damage during carcinogenesis. Oxidative DNA damage, including 7,8-dihydro-8-oxoguanine, play an important role during the cellular transformation. It is also becoming apparent that the unusual activity and subcellular distribution of apurinic/apyrimidinic endonuclease 1, an essential DNA repair factor/redox sensor, affect cancer malignancy by increasing cellular resistance to oxidative stress and by positively influencing cell proliferation. FUTURE DIRECTIONS: Technological advancement in cancer cell biology and genetics has enabled us to monitor the detailed DNA repair activities in the microenvironment. Precise understanding of the intracellular activities of DNA repair proteins for oxidative DNA damage should provide help in understanding how mitochondria, ROS, DNA damage, and repair influence cancer transformation.


Subject(s)
DNA Damage , DNA Repair , Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Humans , Mitochondria/metabolism , Neoplasms/enzymology , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism
15.
Antioxid Redox Signal ; 20(4): 621-39, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23879289

ABSTRACT

SIGNIFICANCE: An emerging concept in DNA repair mechanisms is the evidence that some key enzymes, besides their role in the maintenance of genome stability, display also unexpected noncanonical functions associated with RNA metabolism in specific subcellular districts (e.g., nucleoli). During the evolution of these key enzymes, the acquisition of unfolded domains significantly amplified the possibility to interact with different partners and substrates, possibly explaining their phylogenetic gain of functions. RECENT ADVANCES: After nucleolar stress or DNA damage, many DNA repair proteins can freely relocalize from nucleoli to the nucleoplasm. This process may represent a surveillance mechanism to monitor the synthesis and correct assembly of ribosomal units affecting cell cycle progression or inducing p53-mediated apoptosis or senescence. CRITICAL ISSUES: A paradigm for this kind of regulation is represented by some enzymes of the DNA base excision repair (BER) pathway, such as apurinic/apyrimidinic endonuclease 1 (APE1). In this review, the role of the nucleolus and the noncanonical functions of the APE1 protein are discussed in light of their possible implications in human pathologies. FUTURE DIRECTIONS: A productive cross-talk between DNA repair enzymes and proteins involved in RNA metabolism seems reasonable as the nucleolus is emerging as a dynamic functional hub that coordinates cell growth arrest and DNA repair mechanisms. These findings will drive further analyses on other BER proteins and might imply that nucleic acid processing enzymes are more versatile than originally thought having evolved DNA-targeted functions after a previous life in the early RNA world.


Subject(s)
Cell Nucleolus/enzymology , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Animals , Cell Nucleolus/physiology , DNA Damage , DNA Repair Enzymes/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Nuclear Proteins/metabolism , Nucleophosmin , Protein Structure, Tertiary
16.
Am J Physiol Heart Circ Physiol ; 305(8): H1158-67, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23934858

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that processes DNA-repair function and controls cellular response to oxidative stress. Endothelial progenitor cells (EPCs) are recruited to oxidative stress-rich injured vascular walls and positively contribute to vascular repair and endothelialization. We hypothesized that APE1 functions for EPCs-mediated inhibition of neointima formation in injured vasculature. EPCs isolated from bone marrow cells of C57BL6 mice (12-16 wk old) were able to survive in the presence of hydrogen peroxide (H2O2; up to 1,000 µM) due to the highly expressed reactive oxygen species (ROS) scavengers. However, adhesion capacity of EPCs was significantly inhibited by H2O2 (100 µM) even though an intracellular ROS was retained at small level. An APE1-selective inhibitor or RNA interference-mediated knockdown of endogenous APE1 in EPCs aggravated the H2O2-mediated inhibition of EPCs-adhesion. In contrast, when APE1 was overexpressed in EPCs using an adenovirus harboring the APE1 gene (APE-EPCs), adhesion was significantly improved during oxidative stress. To examine in vivo effects of APE1 in EPCs, APE-EPCs were transplanted via the tail vein after wire-mediated injury of the mouse femoral artery. The number of adherent EPCs at injured vascular walls and the vascular repair effect of EPCs were enhanced in APE-EPCs compared with control EPCs. Among the cellular functions of EPCs, adhesion is especially sensitive to oxidative stress. APE1 enhances in vivo vascular repair effects of EPCs in part through the maintenance of adhesion properties of EPCs. APE1 may be a novel and useful target gene for effective cellular transplantation therapy.


Subject(s)
Cell Adhesion/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Endothelial Cells/physiology , Neointima/physiopathology , Stem Cells/physiology , Animals , Blood Vessels/injuries , Cell Line , Cell Survival , Cells, Cultured , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Endothelial Cells/transplantation , Free Radical Scavengers , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidants/pharmacology , Reactive Oxygen Species
17.
Neoplasma ; 60(4): 384-94, 2013.
Article in English | MEDLINE | ID: mdl-23581410

ABSTRACT

Apurinic/apyrimidinic endonuclease1 (APE1), which has the dual functions of DNA repair and redox regulation, is considered to be a promising potential target in cancer treatment. Microarray and qRT-PCR were used to confirm the change of miRNA followed by analysis with comprehensive bioinformatics-based analysis. Both microarray and qRT-PCR demonstrated that 13 microRNAs (miRNAs) were significantly changed (>2-fold) in APE1 knockdown HOS cells; seven of them (hsa-miR-451, hsa-miR-1290, hsa-miR-765, hsa-miR-483-5p, hsa-miR-513a-5p, hsa-miR-129-5p and hsa-miR-31) were up-regulated and the other six (hsa-miR-29b, hsa-miR-197, has-let-7b, hsa-miR-324-5p, hsa-let-7i and hsa-miR-484) were down-regulated. Furthermore, pathway analysis showed that these miRNAs and their target genes affected by the expression of APE1 were involved in pathways relating to developmental processes, regulation of cellular processes, cell signaling (such as TGF-ß, Wnt, MAPK and the p53 signaling pathway) and cancers. There are putative binding sites of NF-κB, p53, HIF-1α, AP-1, PEBP2, ATF, NF-Y, Pax-2,CREB and c-Myb in the promoters of several down regulated miRNAs, indicating that APE1 may regulate miRNAs via transcription factors. Our data suggest that our understanding of the biological functions of APE1 will inevitably expand due to the novel pathways that APE1 uses to regulate gene expression through miRNAs.


Subject(s)
Bone Neoplasms/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , MicroRNAs/analysis , Osteosarcoma/genetics , Binding Sites , Cell Line, Tumor , Computational Biology , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Down-Regulation , Gene Regulatory Networks , Humans , RNA, Small Interfering/genetics , Transcription Factors/metabolism , Transfection
18.
Toxicology ; 305: 120-9, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23370007

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1)/redox effector factor-1 is a multifunctional enzyme involved in DNA base excision repair and protein redox regulation. Previously, we have showed that lead acetate (Pb) elicits EGFR activation to initiate the SFK/PKCα/Ras/Raf-1/MKK1/2/ERK signaling cascade functioning against genotoxicity. Here, we explore whether APE1 and reactive oxygen species (ROS) affect ERK signaling and cell cycle progression following Pb exposure. We found that Pb induced APE1 expression and ROS generation in CL3 human lung cancer cells. The Pb-elicited ROS levels and cytotoxicity were further enhanced by introducing small interfering RNA specific for APE1 (siAPE1). E3330, an inhibitor of APE1 redox activity, also augmented the ROS levels and cytotoxicity in Pb-treated cells. Intriguingly, the capability of Pb to activate ERK was abolished under siAPE1 or E3330 co-treatments; conversely, forced expression of APE1 up-regulated the ERK activation by Pb or serum in both Cys65-redox activity dependent and independent manners. Moreover, APE1 formed complex with ERK2, and its redox activity could rescue ERK oxidative inactivation. APE1 redox activity also facilitated the Cyclin D1 expression and G1-to-S progression following Pb exposure. In summary, the results indicate that APE1 is a direct redox regulator of ERK for maintaining the kinase activity to promote cell proliferation.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , G1 Phase/drug effects , Mitosis/drug effects , Organometallic Compounds/toxicity , S Phase/drug effects , Signal Transduction/physiology , 3T3 Cells , Animals , Benzoquinones/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Colony-Forming Units Assay , Cysteine/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Humans , Immunoprecipitation , Mice , Plasmids/genetics , Propionates/pharmacology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
19.
Nucleic Acids Res ; 40(21): 10950-63, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22965116

ABSTRACT

Piwi proteins and Piwi-interacting small RNAs (piRNAs) have known functions in transposon silencing in the male germline of fetal and newborn mice. Both are also present in adult testes; however, their function here remains a mystery. Here, we confirm that most piRNAs in meiotic spermatocytes originate from clusters in non-repeat intergenic regions of DNA. The regulation of these piRNA clusters, including the processing of the precursor transcripts into individual piRNAs, is accomplished through mostly unknown processes. We present a possible regulatory mechanism for one such cluster, named cluster 1082B, located on chromosome 7 in the mouse genome. The 1082B precursor transcript and its 788 unique piRNAs are repressed by the Alkbh1 dioxygenase and the testis-specific transcription repressor Tzfp. We observe a remarkable >1000-fold upregulation of individual piRNAs in pachytene spermatocytes isolated from Alkbh1- and Tzfp-deficient murine testes. Repression of cluster 1082B is further supported by the identification of a 10-bp Tzfp recognition sequence contained within the precursor transcript. Downregulation of LINE1 and IAP transcripts in the Alkbh1- and Tzfp-deficient mice leads us to propose a potential role for the 1082B-encoded piRNAs in transposon control.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Gene Expression Regulation , Pachytene Stage/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/physiology , Spermatocytes/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase , Animals , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Down-Regulation , Genes, Intracisternal A-Particle , Long Interspersed Nucleotide Elements , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation , RNA Precursors/metabolism , Repressor Proteins/genetics , Testis/metabolism
20.
Circ Res ; 111(2): 212-9, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22652909

ABSTRACT

RATIONALE: The genetic mechanisms underlying hypertension are unclear, but relative aldosterone excess, present in ≈10% of hypertensive patients, is known to be a heritable trait. This phenotype associates with a T/C single nucleotide polymorphism (SNP) at position -344 of the aldosterone synthase gene (CYP11B2). However, deletion of this SNP has no effect on gene transcription. We have identified another T/C SNP at -1651, in tight linkage disequilibrium with the -344 SNP and here investigate its functional effect on CYP11B2 transcription. OBJECTIVE: We assessed the effect on transcriptional activity of the -1651 T/C SNP in vivo and in vitro and propose the mechanism by which transcriptional activity is altered. METHODS AND RESULTS: We demonstrated that the SNP at -1651 exerts significant allele-dependent effects on CYP11B2 transcription. We confirm binding of the transcriptional repressor APEX1 to -1651T, which is associated with reduced transcriptional activity in relation to the less strongly bound -1651C. We show that inhibiting APEX1 by small molecule inhibition or small interfering RNA (SiRNA) leads to increased CYP11B2 transcription. In addition, overexpression of APEX1 is associated with reduced transcriptional activity. Finally, we also show that -1651T associates with lower excretion rates of aldosterone metabolites in human subjects. CONCLUSIONS: We conclude that APEX1 is a novel transcriptional repressor of CYP11B2 and that differential APEX1 binding at -1651 of CYP11B2 results in altered gene expression. This mechanism may contribute to the observed relationship between CYP11B2 genotype and aldosterone phenotype in a subgroup of hypertensive patients.


Subject(s)
Cytochrome P-450 CYP11B2/biosynthesis , Cytochrome P-450 CYP11B2/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Polymorphism, Single Nucleotide/genetics , Transcription, Genetic/genetics , Adult , Aged , Cytochrome P-450 CYP11B2/antagonists & inhibitors , DNA Repair/genetics , Down-Regulation/genetics , Female , Humans , Male , Middle Aged , Protein Binding/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...