Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 288(18): 12426-36, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23508956

ABSTRACT

Recent evidence suggests a role for base excision repair (BER) proteins in the response to DNA interstrand crosslinks, which block replication and transcription, and lead to cell death and genetic instability. Employing fluorescently tagged fusion proteins and laser microirradiation coupled with confocal microscopy, we observed that the endonuclease VIII-like DNA glycosylase, NEIL1, accumulates at sites of oxidative DNA damage, as well as trioxsalen (psoralen)-induced DNA interstrand crosslinks, but not to angelicin monoadducts. While recruitment to the oxidative DNA lesions was abrogated by the anti-oxidant N-acetylcysteine, this treatment did not alter the accumulation of NEIL1 at sites of interstrand crosslinks, suggesting distinct recognition mechanisms. Consistent with this conclusion, recruitment of the NEIL1 population variants, G83D, C136R, and E181K, to oxidative DNA damage and psoralen-induced interstrand crosslinks was differentially affected by the mutation. NEIL1 recruitment to psoralen crosslinks was independent of the nucleotide excision repair recognition factor, XPC. Knockdown of NEIL1 in LN428 glioblastoma cells resulted in enhanced recruitment of XPC, a more rapid removal of digoxigenin-tagged psoralen adducts, and decreased cellular sensitivity to trioxsalen plus UVA, implying that NEIL1 and BER may interfere with normal cellular processing of interstrand crosslinks. While exhibiting no enzymatic activity, purified NEIL1 protein bound stably to psoralen interstrand crosslink-containing synthetic oligonucleotide substrates in vitro. Our results indicate that NEIL1 recognizes specifically and distinctly interstrand crosslinks in DNA, and can obstruct the efficient removal of lethal crosslink adducts.


Subject(s)
Cross-Linking Reagents/pharmacology , DNA Adducts/metabolism , DNA Damage , DNA Glycosylases/metabolism , DNA Repair/drug effects , Ficusin/pharmacology , Acetylcysteine/pharmacology , DNA Adducts/genetics , DNA Glycosylases/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Free Radical Scavengers/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , Oxidation-Reduction/drug effects , Protein Binding/drug effects , Protein Binding/radiation effects , Ultraviolet Rays/adverse effects
2.
PLoS One ; 7(10): e47974, 2012.
Article in English | MEDLINE | ID: mdl-23110144

ABSTRACT

The major human apurinic/apyrimidinic endonuclease APE1 plays a pivotal role in the repair of base damage via participation in the DNA base excision repair (BER) pathway. Increased activity of APE1, often observed in tumor cells, is thought to contribute to resistance to various anticancer drugs, whereas down-regulation of APE1 sensitizes cells to DNA damaging agents. Thus, inhibiting APE1 repair endonuclease function in cancer cells is considered a promising strategy to overcome therapeutic agent resistance. Despite ongoing efforts, inhibitors of APE1 with adequate drug-like properties have yet to be discovered. Using a kinetic fluorescence assay, we conducted a fully-automated high-throughput screen (HTS) of the NIH Molecular Libraries Small Molecule Repository (MLSMR), as well as additional public collections, with each compound tested as a 7-concentration series in a 4 µL reaction volume. Actives identified from the screen were subjected to a panel of confirmatory and counterscreen tests. Several active molecules were identified that inhibited APE1 in two independent assay formats and exhibited potentiation of the genotoxic effect of methyl methanesulfonate with a concomitant increase in AP sites, a hallmark of intracellular APE1 inhibition; a number of these chemotypes could be good starting points for further medicinal chemistry optimization. To our knowledge, this represents the largest-scale HTS to identify inhibitors of APE1, and provides a key first step in the development of novel agents targeting BER for cancer treatment.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Small Molecule Libraries , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Methyl Methanesulfonate/antagonists & inhibitors , Methyl Methanesulfonate/pharmacology , Molecular Structure , Structure-Activity Relationship
3.
J Med Chem ; 55(7): 3101-12, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22455312

ABSTRACT

APE1 is an essential protein that operates in the base excision repair (BER) pathway and is responsible for ≥95% of the total apurinic/apyrimidinic (AP) endonuclease activity in human cells. BER is a major pathway that copes with DNA damage induced by several anticancer agents, including ionizing radiation and temozolomide. Overexpression of APE1 and enhanced AP endonuclease activity have been linked to increased resistance of tumor cells to treatment with monofunctional alkylators, implicating inhibition of APE1 as a valid strategy for cancer therapy. We report herein the results of a focused medicinal chemistry effort around a novel APE1 inhibitor, N-(3-(benzo[d]thiazol-2-yl)-6-isopropyl-4,5,6,7-tetrahydrothieno[2,3-c]pyridin-2-yl)acetamide (3). Compound 3 and related analogues exhibit single-digit micromolar activity against the purified APE1 enzyme and comparable activity in HeLa whole cell extract assays and potentiate the cytotoxicity of the alkylating agents methylmethane sulfonate and temozolomide. Moreover, this class of compounds possesses a generally favorable in vitro ADME profile, along with good exposure levels in plasma and brain following intraperitoneal dosing (30 mg/kg body weight) in mice.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzothiazoles/chemical synthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Thiophenes/chemical synthesis , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Benzothiazoles/pharmacokinetics , Benzothiazoles/pharmacology , Brain/metabolism , Cell Extracts , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Synergism , HeLa Cells , Humans , Injections, Intraperitoneal , Methyl Methanesulfonate/pharmacology , Mice , Small Molecule Libraries , Structure-Activity Relationship , Temozolomide , Thiophenes/pharmacokinetics , Thiophenes/pharmacology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...