Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38645048

ABSTRACT

The multitude of DNA lesion types, and the nuclear dynamic context in which they occur, present a challenge for genome integrity maintenance as this requires the engagement of different DNA repair pathways. Specific 'repair controllers' that facilitate DNA repair pathway crosstalk between double strand break (DSB) repair and base excision repair (BER), and regulate BER protein trafficking at lesion sites, have yet to be identified. We find that DNA polymerase ß (Polß), crucial for BER, is ubiquitylated in a BER complex-dependent manner by TRIP12, an E3 ligase that partners with UBR5 and restrains DSB repair signaling. Here we find that, TRIP12, but not UBR5, controls cellular levels and chromatin loading of Polß. Required for Polß foci formation, TRIP12 regulates Polß involvement after DNA damage. Notably, excessive TRIP12-mediated shuttling of Polß affects DSB formation and radiation sensitivity, underscoring its precedence for BER. We conclude that the herein discovered trafficking function at the nexus of DNA repair signaling pathways, towards Polß-directed BER, optimizes DNA repair pathway choice at complex lesion sites.

2.
Cancers (Basel) ; 16(2)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38254819

ABSTRACT

O6-methylguanine-DNA methyltransferase (MGMT or AGT) is a DNA repair protein with the capability to remove alkyl groups from O6-AlkylG adducts. Moreover, MGMT plays a crucial role in repairing DNA damage induced by methylating agents like temozolomide and chloroethylating agents such as carmustine, and thereby contributes to chemotherapeutic resistance when these agents are used. This review delves into the structural roles and repair mechanisms of MGMT, with emphasis on the potential structural and functional roles of the N-terminal domain of MGMT. It also explores the development of cancer therapeutic strategies that target MGMT. Finally, it discusses the intriguing crosstalk between MGMT and other DNA repair pathways.

3.
Cell Rep ; 37(5): 109917, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731617

ABSTRACT

Assembly and disassembly of DNA repair protein complexes at DNA damage sites are essential for maintaining genomic integrity. Investigating factors coordinating assembly of the base excision repair (BER) proteins DNA polymerase ß (Polß) and XRCC1 to DNA lesion sites identifies a role for Polß in regulating XRCC1 disassembly from DNA repair complexes and, conversely, demonstrates Polß's dependence on XRCC1 for complex assembly. LivePAR, a genetically encoded probe for live-cell imaging of poly(ADP-ribose) (PAR), reveals that Polß and XRCC1 require PAR for repair-complex assembly, with PARP1 and PARP2 playing unique roles in complex dynamics. Further, BER complex assembly is modulated by attenuation/augmentation of NAD+ biosynthesis. Finally, SIRT6 does not modulate PARP1 or PARP2 activation but does regulate XRCC1 recruitment, leading to diminished Polß abundance at sites of DNA damage. These findings highlight coordinated yet independent roles for PARP1, PARP2, and SIRT6 and their regulation by NAD+ bioavailability to facilitate BER.


Subject(s)
DNA Breaks, Single-Stranded , DNA Repair , DNA, Neoplasm/metabolism , NAD/metabolism , Neoplasms/enzymology , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Sirtuins/metabolism , A549 Cells , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , DNA, Neoplasm/genetics , Humans , Kinetics , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , Sirtuins/genetics , X-ray Repair Cross Complementing Protein 1/genetics , X-ray Repair Cross Complementing Protein 1/metabolism
4.
Nucleic Acids Res ; 47(12): 6269-6286, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31287140

ABSTRACT

Protein-protein interactions regulate many essential enzymatic processes in the cell. Somatic mutations outside of an enzyme active site can therefore impact cellular function by disruption of critical protein-protein interactions. In our investigation of the cellular impact of the T304I cancer mutation of DNA Polymerase ß (Polß), we find that mutation of this surface threonine residue impacts critical Polß protein-protein interactions. We show that proteasome-mediated degradation of Polß is regulated by both ubiquitin-dependent and ubiquitin-independent processes via unique protein-protein interactions. The ubiquitin-independent proteasome pathway regulates the stability of Polß in the cytosol via interaction between Polß and NAD(P)H quinone dehydrogenase 1 (NQO1) in an NADH-dependent manner. Conversely, the interaction of Polß with the scaffold protein X-ray repair cross complementing 1 (XRCC1) plays a role in the localization of Polß to the nuclear compartment and regulates the stability of Polß via a ubiquitin-dependent pathway. Further, we find that oxidative stress promotes the dissociation of the Polß/NQO1 complex, enhancing the interaction of Polß with XRCC1. Our results reveal that somatic mutations such as T304I in Polß impact critical protein-protein interactions, altering the stability and sub-cellular localization of Polß and providing mechanistic insight into how key protein-protein interactions regulate cellular responses to stress.


Subject(s)
DNA Polymerase beta/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress , X-ray Repair Cross Complementing Protein 1/metabolism , Cell Line, Tumor , Chromatin/enzymology , Colonic Neoplasms/genetics , DNA Polymerase beta/chemistry , DNA Polymerase beta/genetics , Enzyme Stability , Humans , Mutation , NAD/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitination
5.
Proc Natl Acad Sci U S A ; 112(28): 8602-7, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26124145

ABSTRACT

Free radical attack on the C1' position of DNA deoxyribose generates the oxidized abasic (AP) site 2-deoxyribonolactone (dL). Upon encountering dL, AP lyase enzymes such as DNA polymerase ß (Polß) form dead-end, covalent intermediates in vitro during attempted DNA repair. However, the conditions that lead to the in vivo formation of such DNA-protein cross-links (DPC), and their impact on cellular functions, have remained unknown. We adapted an immuno-slot blot approach to detect oxidative Polß-DPC in vivo. Treatment of mammalian cells with genotoxic oxidants that generate dL in DNA led to the formation of Polß-DPC in vivo. In a dose-dependent fashion, Polß-DPC were detected in MDA-MB-231 human cells treated with the antitumor drug tirapazamine (TPZ; much more Polß-DPC under 1% O2 than under 21% O2) and even more robustly with the "chemical nuclease" 1,10-copper-ortho-phenanthroline, Cu(OP)2. Mouse embryonic fibroblasts challenged with TPZ or Cu(OP)2 also incurred Polß-DPC. Nonoxidative agents did not generate Polß-DPC. The cross-linking in vivo was clearly a result of the base excision DNA repair pathway: oxidative Polß-DPC depended on the Ape1 AP endonuclease, which generates the Polß lyase substrate, and they required the essential lysine-72 in the Polß lyase active site. Oxidative Polß-DPC had an unexpectedly short half-life (∼ 30 min) in both human and mouse cells, and their removal was dependent on the proteasome. Proteasome inhibition under Cu(OP)2 treatment was significantly more cytotoxic to cells expressing wild-type Polß than to cells with the lyase-defective form. That observation underscores the genotoxic potential of oxidative Polß-DPC and the biological pressure to repair them.


Subject(s)
DNA Damage , DNA Polymerase beta/metabolism , DNA/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Oxidation-Reduction
6.
Nat Commun ; 5: 5513, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25423885

ABSTRACT

Cellular DNA repair processes are crucial to maintain genome stability and integrity. In DNA base excision repair, a tight heterodimer complex formed by DNA polymerase ß (Polß) and XRCC1 is thought to facilitate repair by recruiting Polß to DNA damage sites. Here we show that disruption of the complex does not impact DNA damage response or DNA repair. Instead, the heterodimer formation is required to prevent ubiquitylation and degradation of Polß. In contrast, the stability of the XRCC1 monomer is protected from CHIP-mediated ubiquitylation by interaction with the binding partner HSP90. In response to cellular proliferation and DNA damage, proteasome and HSP90-mediated regulation of Polß and XRCC1 alters the DNA repair complex architecture. We propose that protein stability, mediated by DNA repair protein complex formation, functions as a regulatory mechanism for DNA repair pathway choice in the context of cell cycle progression and genome surveillance.


Subject(s)
DNA Polymerase beta/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Cell Line , DNA Damage , DNA Polymerase beta/genetics , DNA-Binding Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Humans , Models, Molecular , Protein Binding , X-ray Repair Cross Complementing Protein 1
7.
Mutat Res ; 741-742: 1-10, 2013.
Article in English | MEDLINE | ID: mdl-23500083

ABSTRACT

Bis-electrophiles including dibromoethane and epibromohydrin can react with O(6)-alkylguanine-DNA alkyltransferase (AGT) and form AGT-DNA crosslinks in vitro and in vivo. The presence of human AGT (hAGT) paradoxically increases the mutagenicity and cytotoxicity of bis-electrophiles in cells. Here we establish a bacterial system to study the repair mechanism and cellular responses to DNA-protein crosslinks (DPCs) in vivo. Results show that both nucleotide excision repair (NER) and homologous recombination (HR) pathways can process hAGT-DNA crosslinks with HR playing a dominant role. Mutation spectra show that HR has no strand preference but NER favors processing of the DPCs in the transcribed strand; UvrA, UvrB and Mfd can interfere with small size DPCs but only UvrA can interfere with large size DPCs in the transcribed strand processed by HR. Further, we found that DPCs at TA deoxynucleotide sites are very inefficiently processed by NER and the presence of NER can interfere with these DNA lesions processed by HR. These data indicate that NER and HR can process DPCs cooperatively and competitively and NER processes DPCs with base and strand preference. Therefore, the formation of hAGT-DNA crosslinks can be a plausible and specific system to study the repair mechanism and effects of DPCs precisely in vivo.


Subject(s)
Cross-Linking Reagents , DNA Repair/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Homologous Recombination , DNA Damage , DNA, Bacterial/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Humans , Mutation/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Recombination, Genetic
8.
PLoS One ; 6(3): e17862, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21448280

ABSTRACT

An siRNA screen has identified several proteins throughout the base excision repair (BER) pathway of oxidative DNA damage as important for efficient HIV infection. The proteins identified included early repair factors such as the base damage recognition glycosylases OGG1 and MYH and the late repair factor POLß, implicating the entire BER pathway. Murine cells with deletions of the genes Ogg1, Myh, Neil1 and Polß recapitulate the defect of HIV infection in the absence of BER. Defective infection in the absence of BER proteins was also seen with the lentivirus FIV, but not the gammaretrovirus MMLV. BER proteins do not affect HIV infection through its accessory genes nor the central polypurine tract. HIV reverse transcription and nuclear entry appear unaffected by the absence of BER proteins. However, HIV integration to the host chromosome is reduced in the absence of BER proteins. Pre-integration complexes from BER deficient cell lines show reduced integration activity in vitro. Integration activity is restored by addition of recombinant BER protein POLß. Lentiviral infection and integration efficiency appears to depend on the presence of BER proteins.


Subject(s)
DNA Repair/genetics , Lentivirus/physiology , Signal Transduction/genetics , Virus Integration/genetics , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Nucleus/metabolism , Cell Survival , DNA Damage , DNA, Complementary/genetics , DNA, Viral/genetics , Gene Deletion , HIV Infections/genetics , Humans , Lentivirus Infections/genetics , Mice , Reverse Transcription/genetics , Time Factors
9.
Org Biomol Chem ; 8(19): 4414-26, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20714665

ABSTRACT

O(6)-2'-Deoxyguanosine-alkyl-O(6)-2'-deoxyguanosine interstrand DNA cross-links (ICLs) with a four and seven methylene linkage in a 5'-GNC- motif have been synthesized and their repair by human O6-alkylguanine-DNA alkyltransferase (hAGT) investigated. Duplexes containing 11 base-pairs with the ICLs in the center were assembled by automated DNA solid-phase synthesis using a cross-linked 2'-deoxyguanosine dimer phosphoramidite, prepared via a seven step synthesis which employed the Mitsunobu reaction to introduce the alkyl lesion at the O(6) atom of guanine. Introduction of the four and seven carbon ICLs resulted in no change in duplex stability based on UV thermal denaturation experiments compared to a non-cross-linked control. Circular dichroism spectra of these ICL duplexes exhibited features of a B-form duplex, similar to the control, suggesting that these lesions induce little overall change in structure. The efficiency of repair by hAGT was examined and it was shown that hAGT repairs both ICL containing duplexes, with the heptyl ICL repaired more efficiently relative to the butyl cross-link. These results were reproducible with various hAGT mutants including one that contains a novel V148L mutation. The ICL duplexes displayed similar binding affinities to a C145S hAGT mutant compared to the unmodified duplex with the seven carbon containing ICLs displaying slightly higher binding. Experiments with CHO cells to investigate the sensitivity of these cells to busulfan and hepsulfam demonstrate that hAGT reduces the cytotoxicity of hepsulfam suggesting that the O(6)-2'-deoxyguanosine-alkyl-O(6)-2'-deoxyguanosine interstrand DNA cross-link may account for at least part of the cytotoxicity of this agent.


Subject(s)
DNA Repair , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylating Agents/pharmacology , Animals , Busulfan/pharmacology , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , DNA/chemistry , DNA/metabolism , Deoxyguanosine/chemical synthesis , Humans , Mutation , Nucleic Acid Denaturation , O(6)-Methylguanine-DNA Methyltransferase/genetics , Sulfonic Acids/pharmacology
10.
J Biol Chem ; 285(11): 8185-95, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20026607

ABSTRACT

O(6)-Alkylguanine-DNA alkyltransferase (AGT) plays a major role in repair of the cytotoxic and mutagenic lesion O(6)-methylguanine (m(6)G) in DNA. Unlike the Escherichia coli alkyltransferase Ogt that also repairs O(4)-methylthymine (m(4)T) efficiently, the human AGT (hAGT) acts poorly on m(4)T. Here we made several hAGT mutants in which residues near the cysteine acceptor site were replaced by corresponding residues from Ogt to investigate the basis for the inefficiency of hAGT in repair of m(4)T. Construct hAGT-03 (where hAGT sequence -V(149)CSSGAVGN(157)- was replaced with the corresponding Ogt -I(143)GRNGTMTG(151)-) exhibited enhanced m(4)T repair activity in vitro compared with hAGT. Three AGT proteins (hAGT, hAGT-03, and Ogt) exhibited similar protection from killing by N-methyl-N'-nitro-N-nitrosoguanidine and caused a reduction in m(6)G-induced G:C to A:T mutations in both nucleotide excision repair (NER)-proficient and -deficient Escherichia coli strains that lack endogenous AGTs. hAGT-03 resembled Ogt in totally reducing the m(4)T-induced T:A to C:G mutations in NER-proficient and -deficient strains. Surprisingly, wild type hAGT expression caused a significant but incomplete decrease in NER-deficient strains but a slight increase in T:A to C:G mutation frequency in NER-proficient strains. The T:A to C:G mutations due to O(4)-alkylthymine formed by ethylating and propylating agents were also efficiently reduced by either hAGT-03 or Ogt, whereas hAGT had little effect irrespective of NER status. These results show that specific alterations in the hAGT active site facilitate efficient recognition and repair of O(4)-alkylthymines and reveal damage-dependent interactions of base and nucleotide excision repair.


Subject(s)
DNA Adducts/metabolism , DNA Repair/physiology , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Thymine/metabolism , Alkylation/physiology , Catalytic Domain , DNA Damage/physiology , Escherichia coli/genetics , Ethane/metabolism , Humans , Methane/metabolism , Mutagenesis/physiology , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Propane/metabolism , Protein Structure, Tertiary
11.
J Biol Chem ; 284(34): 22601-10, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19531487

ABSTRACT

O(6)-alkyldeoxyguanine adducts induced by tobacco-specific nitrosamines are repaired by O(6)-alkylguanine DNA alkyltransferase (AGT), which transfers the O(6)-alkyl group from the damaged base to a cysteine residue within the protein. In the present study, a mass spectrometry-based approach was used to analyze the effects of cytosine methylation on the kinetics of AGT repair of O(6)-methyldeoxyguanosine (O(6)-Me-dG) adducts placed within frequently mutated 5'-CG-3' dinucleotides of the p53 tumor suppressor gene. O(6)-Me-dG-containing DNA duplexes were incubated with human recombinant AGT protein, followed by rapid quenching, acid hydrolysis, and isotope dilution high pressure liquid chromatography-electrospray ionization tandem mass spectrometry analysis of unrepaired O(6)-methylguanine. Second-order rate constants were calculated in the absence or presence of the C-5 methyl group at neighboring cytosine residues. We found that the kinetics of AGT-mediated repair of O(6)-Me-dG were affected by neighboring 5-methylcytosine ((Me)C) in a sequence-dependent manner. AGT repair of O(6)-Me-dG adducts placed within 5'-CG-3' dinucleotides of p53 codons 245 and 248 was hindered when (Me)C was present in both DNA strands. In contrast, cytosine methylation within p53 codon 158 slightly increased the rate of O(6)-Me-dG repair by AGT. The effects of (Me)C located immediately 5' and in the base paired position to O(6)-Me-dG were not additive as revealed by experiments with hypomethylated sequences. Furthermore, differences in dealkylation rates did not correlate with AGT protein affinity for cytosine-methylated and unmethylated DNA duplexes or with the rates of AGT-mediated nucleotide flipping, suggesting that (Me)C influences other kinetic steps involved in repair, e.g. the rate of alkyl transfer from DNA to AGT.


Subject(s)
Cytosine/metabolism , DNA Repair/physiology , Dinucleoside Phosphates/metabolism , Guanine/analogs & derivatives , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Chromatography, High Pressure Liquid , Circular Dichroism , Codon/genetics , DNA Methylation , DNA Repair/genetics , Dinucleoside Phosphates/genetics , Electrophoretic Mobility Shift Assay , Genes, p53/genetics , Guanine/metabolism , Humans , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Ultraviolet Rays
12.
Biochemistry ; 47(50): 13404-17, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19053272

ABSTRACT

Putrescine (1,4-diaminobutane) activates the autoprocessing and decarboxylation reactions of human S-adenosylmethionine decarboxylase (AdoMetDC), a critical enzyme in the polyamine biosynthetic pathway. In human AdoMetDC, putrescine binds in a buried pocket containing acidic residues Asp174, Glu178, and Glu256. The pocket is away from the active site but near the dimer interface; however, a series of hydrophilic residues connect the putrescine binding site and the active site. Mutation of these acidic residues modulates the effects of putrescine. D174N, E178Q, and E256Q mutants were expressed and dialyzed to remove putrescine and studied biochemically using X-ray crystallography, UV-CD spectroscopy, analytical ultracentrifugation, and ITC binding studies. The results show that the binding of putrescine to the wild type dimeric protein is cooperative. The D174N mutant does not bind putrescine, and the E178Q and E256Q mutants bind putrescine weakly with no cooperativity. The crystal structure of the mutants with and without putrescine and their complexes with S-adenosylmethionine methyl ester were obtained. Binding of putrescine results in a reorganization of four aromatic residues (Phe285, Phe315, Tyr318, and Phe320) and a conformational change in the loop 312-320. The loop shields putrescine from the external solvent, enhancing its electrostatic and hydrogen bonding effects. The E256Q mutant with putrescine added shows an alternate conformation of His243, Glu11, Lys80, and Ser229, the residues that link the active site and the putrescine binding site, suggesting that putrescine activates the enzyme through electrostatic effects and acts as a switch to correctly orient key catalytic residues.


Subject(s)
Adenosylmethionine Decarboxylase/chemistry , Adenosylmethionine Decarboxylase/metabolism , Putrescine/chemistry , Putrescine/metabolism , Crystallography, X-Ray , Dimerization , Enzyme Activation/physiology , Humans , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Static Electricity
13.
J Med Chem ; 51(22): 7144-53, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18973327

ABSTRACT

O(6)-Benzylguanine is an irreversible inactivator of O(6)-alkylguanine-DNA alkyltransferase currently in clinical trials to overcome alkyltransferase-mediated resistance to certain cancer chemotherapeutic alkylating agents. In order to produce more soluble alkyltransferase inhibitors, we have synthesized three aminomethyl-substituted O(6)-benzylguanines and the three methyl analogs and found that the substitution of aminomethyl at the meta-position greatly enhances inactivation of alkyltransferase, whereas para-substitution has little effect and ortho-substitution virtually eliminates activity. Molecular modeling of their interactions with alkyltransferase provided a molecular explanation for these results. The square of the correlation coefficient (R(2)) obtained between E-model scores (obtained from GLIDE XP/QPLD docking calculations) vs log(ED(50)) values via a linear regression analysis was 0.96. The models indicate that the ortho-substitution causes a steric clash interfering with binding, whereas the meta-aminomethyl substitution allows an interaction of the amino group to generate an additional hydrogen bond with the protein.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Computer Simulation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guanine/chemical synthesis , Guanine/chemistry , Guanine/pharmacology , Humans , Hydrogen Bonding , Ligands , Models, Chemical , Models, Molecular , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
14.
Biochemistry ; 47(41): 10892-903, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18803403

ABSTRACT

O (6)-Alkylguanine-DNA alkyltransferase (AGT) plays an important role by protecting cells from alkylating agents. This reduces the frequency of carcinogenesis and mutagenesis initiated by such agents, but AGT also provides a major resistance mechanism to some chemotherapeutic drugs. To improve our understanding of the AGT-mediated repair reaction and our understanding of the spectrum of repairable damage, we have studied the ability of AGT to repair interstrand cross-link DNA damage where the two DNA strands are joined via the guanine- O (6) in each strand. An oligodeoxyribonucleotide containing a heptane cross-link was repaired with initial formation of an AGT-oligo complex and further reaction of a second AGT molecule yielding a hAGT dimer and free oligo. However, an oligodeoxyribonucleotide with a butane cross-link was a very poor substrate for AGT-mediated repair, and only the first reaction that forms an AGT-oligo complex could be detected. Models of the reaction of these substrates in the AGT active site show that the DNA duplex is forced apart locally to repair the first guanine. This reaction is greatly hindered with the butane cross-link, which is mostly buried in the active site pocket and limited in conformational flexibility. This limitation also prevents the adoption of a conformation for the second reaction to repair the AGT-oligo complex. These results are consistent with the postulated mechanism of AGT repair that involves DNA binding and flipping of the substrate nucleotide and indicate that hAGT can repair some types of interstrand cross-link damage.


Subject(s)
DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Tumor Suppressor Proteins/metabolism , Blotting, Western , Chromatography, High Pressure Liquid , DNA Damage , DNA Modification Methylases/chemistry , DNA Repair Enzymes/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Models, Molecular , Tumor Suppressor Proteins/chemistry
15.
Chem Res Toxicol ; 21(4): 787-95, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18324787

ABSTRACT

The antitumor activity of chemotherapeutic nitrogen mustards including chlorambucil, cyclophosphamide, and melphalan is commonly attributed to their ability to induce DNA-DNA cross-links by consecutive alkylation of two nucleophilic sites within the DNA duplex. DNA-protein cross-linking by nitrogen mustards is not well characterized, probably because of its inherent complexity and the insufficient sensitivity of previous methodologies. If formed, DNA-protein conjugates are likely to contribute to both target and off-target cytotoxicity of nitrogen mustard drugs. Here, we show that the DNA repair protein, O (6)-alkylguanine DNA alkyltransferase (AGT), can be readily cross-linked to DNA in the presence of nitrogen mustards. Both chlorambucil and mechlorethamine induced the formation of covalent conjugates between (32)P-labeled double-stranded oligodeoxynucleotides and recombinant human AGT protein, which were detected by SDS-PAGE. Capillary HPLC-electrospray ionization mass spectrometry (ESI-MS) analysis of AGT that had been treated with the guanine half-mustards of chlorambucil or mechlorethamine revealed the ability of the protein to form either one or two cross-links to guanine. C145A AGT (a variant containing a single point mutation in the protein's active site) was found capable of forming a single guanine conjugate, while cross-linking was virtually abolished upon treatment of the C145A/C150S AGT double mutant with the guanine half-mustards. HPLC-ESI (+)-MS/MS sequencing of tryptic peptides obtained from the wild-type AGT protein that had been treated with nitrogen mustards in the presence of DNA confirmed that the cross-linking took place between the N7 position of guanine in DNA and two active site residues within the AGT protein (Cys (145) and Cys (150)). The exact chemical structures of AGT-DNA cross-links induced by chlorambucil and mechlorethamine were identified as N-(2-[ S-cysteinyl]ethyl)- N-(2-[guan-7-yl]ethyl)- p-aminophenylbuyric acid and N-(2-[ S-cysteinyl]ethyl)- N-(2-[guan-7-yl]ethyl)methylamine, respectively, based upon HPLC-MS/MS analysis of protein hydrolysates in parallel with the corresponding amino acid conjugates prepared synthetically. Mechlorethamine-induced AGT-DNA conjugates were isolated from protein extracts of AGT-expressing CHO cells but not control cells, demonstrating that nitrogen mustards can cross-link the AGT protein to DNA in the presence of other nuclear proteins. Because AGT is overexpressed in many tumor types, further investigations of the potential role of AGT-DNA cross-linking in the antitumor and mutagenic activity of antitumor nitrogen mustards are warranted.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/metabolism , Nitrogen Mustard Compounds/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , DNA Repair
16.
Biochem Pharmacol ; 75(3): 618-26, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17996846

ABSTRACT

The human DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (hAGT) is an important source of resistance to some therapeutic alkylating agents and attempts to circumvent this resistance by the use of hAGT inhibitors have reached clinical trials. Several human polymorphisms in the MGMT gene that encodes hAGT have been described including L84F and the linked double alteration I143V/K178R. We have investigated the inactivation of these variants and the much rarer variant W65C by O(6)-benzylguanine, which is currently in clinical trials, and a number of other second generation hAGT inhibitors that contain folate derivatives (O(4)-benzylfolic acid, the 3' and 5' folate esters of O(6)-benzyl-2'-deoxyguanosine and the folic acid gamma ester of O(6)-(p-hydroxymethyl)benzylguanine). The I143V/K178R variant was resistant to all of these compounds. The resistance was due solely to the I143V change. These results suggest that the frequency of the I143V/K178R variant among patients in the clinical trials with hAGT inhibitors and the correlation with response should be considered.


Subject(s)
Enzyme Inhibitors/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Clinical Trials as Topic , Humans , O(6)-Methylguanine-DNA Methyltransferase/genetics , Polymorphism, Genetic
17.
J Med Chem ; 50(21): 5193-201, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17880193

ABSTRACT

O6-Alkylguanine-DNA alkyltransferase (alkyltransferase) provides an important source of resistance to some cancer chemotherapeutic alkylating agents. Folate ester derivatives of O6-benzyl-2'-deoxyguanosine and of O6-[4-(hydroxymethyl)benzyl]guanine were synthesized and tested for their ability to inactivate human alkyltransferase. Inactivation of alkyltransferase by the gamma-folate ester of O6-[4-(hydroxymethyl)benzyl]guanine was similar to that of the parent base. The gamma-folate esters of O6-benzyl-2'-deoxyguanosine were more potent alkyltransferase inactivators than the parent nucleoside. The 3'-ester was considerably more potent than the 5'-ester and was more than an order of magnitude more active than O6-benzylguanine, which is currently in clinical trials to enhance therapy with alkylating agents. They were also able to sensitize human tumor cells to killing by 1,3-bis(2-chloroethyl)-1-nitrosourea, with O6-benzyl-3'-O-(gamma-folyl)-2'-deoxyguanosine being most active. These compounds provide a new class of highly water-soluble alkyltransferase inactivators and form the basis to construct more tumor-specific and potent compounds targeting this DNA repair protein.


Subject(s)
Deoxyguanosine/analogs & derivatives , Folic Acid/analogs & derivatives , Folic Acid/chemical synthesis , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Carrier Proteins/metabolism , Cell Line, Tumor , Deoxyguanosine/chemical synthesis , Deoxyguanosine/pharmacology , Drug Resistance, Neoplasm , Esters , Folate Receptors, GPI-Anchored , Folic Acid/pharmacology , Guanine/chemical synthesis , Guanine/pharmacology , Humans , Hydrolysis , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Receptors, Cell Surface/metabolism , Solubility , Structure-Activity Relationship
18.
Mutat Res ; 624(1-2): 71-9, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17521682

ABSTRACT

Intake of linoleic acid (LA) increased etheno-DNA adducts induced by lipid peroxidation (LPO) in white blood cells (WBC) of female but not of male volunteers [J. Nair, C.E. Vaca, I. Velic, M. Mutanen, L.M. Valsta, H. Bartsch, High dietary omega-6 polyunsaturated fatty acids drastically increase the formation of etheno-DNA adducts in white blood cells of female subjects, Cancer Epidemiol. Biomarkers Prev. 6 (1997) 597-601]. Etheno-adducts were measured in rats gavaged with LA, oleic acid (OA) and saturated fatty acid rich coconut oil for 30 days. DNA from organs and total WBC was analyzed for 1, N(6)-ethenodeoxyadenosine (varepsilondA) and 3, N(4)-ethenodeoxycytidine (varepsilondC) by immunoaffinity/(32)P-postlabeling. Colon was the most affected target with LA-treatment, where etheno-adducts were significantly elevated in both sexes. In WBC both adducts were elevated only in LA-treated females. Unexpectedly, OA treatment enhanced etheno-adduct levels in prostate 3-9 fold. Our results in rodents confirm the gender-specific increase of etheno-adducts in WBC-DNA, likely due to LPO induced by redox-cycling of 4-hydroxyestradiol. Colon was a target for LPO-derived DNA-adducts in both LA-treated male and female rats, supporting their role in omega-6 PUFA induced colon carcinogenesis.


Subject(s)
DNA Adducts/biosynthesis , Linoleic Acid/toxicity , Oleic Acid/toxicity , Plant Oils/toxicity , Animals , Cattle , Coconut Oil , Colon/drug effects , Colon/metabolism , Colonic Neoplasms/etiology , DNA Adducts/analysis , Deoxyadenosines/analysis , Deoxycytidine/analogs & derivatives , Deoxycytidine/analysis , Female , Humans , In Vitro Techniques , Leukocytes/drug effects , Leukocytes/metabolism , Linoleic Acid/administration & dosage , Lipid Peroxidation/drug effects , Male , Oleic Acid/administration & dosage , Organ Specificity , Plant Oils/administration & dosage , Rats , Sex Characteristics
19.
DNA Repair (Amst) ; 6(8): 1071-8, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17482892

ABSTRACT

This article summarizes the current understanding of known variant forms of the MGMT gene that encode an altered protein. Epidemiological studies have been carried out to test whether these alterations are associated with altered cancer risk. Laboratory studies using recombinant proteins and cells expressing the known variants have investigated the possible effects of these sequence alterations on the ability of the encoded O(6)-alkylguanine-DNA alkyltransferase protein to protect cells from alkylation damage and to respond to therapeutic inactivators currently undergoing trials for cancer chemotherapy.


Subject(s)
DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Genetic Variation , O(6)-Methylguanine-DNA Methyltransferase/genetics , Tumor Suppressor Proteins/genetics , Amino Acid Substitution , DNA Modification Methylases/metabolism , DNA Repair , DNA Repair Enzymes/metabolism , Female , Gene Expression , Humans , In Vitro Techniques , Male , Neoplasms/enzymology , Neoplasms/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Polymorphism, Single Nucleotide , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/metabolism
20.
Chem Res Toxicol ; 20(1): 155-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17226938

ABSTRACT

The production of mutations and the reduction in survival of cells treated with alpha,omega-dihaloalkanes is greatly enhanced by the presence of O6-alkylguanine-DNA alkyltransferase (AGT), a DNA repair protein that removes O6-alkylguanine adducts from DNA [Liu, L., Hachey, D. L., Valadez, G., Williams, K. M., Guengerich, F. P., Loktionova, N. A., Kanugula, S., and Pegg, A. E. (2004) J. Biol. Chem. 279, 4250-4259]. The effects of alterations to key residues in the active site of AGT were studied using AGTs with point mutations. It was found that mutants of AGT at positions Tyr114, Arg128, Pro140, Gly156, Gly160, and Tyr158 did not bring about the increase in genotoxicity of 1,2-dibromoethane seen with wild-type AGT, although these mutants, with the exception of those at Tyr114 and Arg128, are known to have sufficient AGT repair function to be able to protect cells from alkylating agents. The R128A mutant was able to react with 1,2-dibromoethane at the Cys145 acceptor site, but the resulting AGT-Cys145S-(CH2)2Br was much less able to produce a covalent adduct with DNA. This result is explained by the need for AGT to induce a structural change in the DNA "flipping" of a guanine nucleotide into the substrate binding pocket where Cys145 is located since the side chain of residue Arg128 plays a critical role in this reaction. Point mutations in AGT at the other sites (Y114A, P140K, and Y158H) reduced the ability of the protein to react with 1,2-dibromoethane as measured by the loss of activity. These results were confirmed by MS analysis of the tryptic peptide that contains the modified Cys145. There was no change in the stability of the AGT-Cys145S-(CH2)2Br intermediate formed in mutants Y158H and P140K. The reaction was studied in detail with mutant P140K using dihaloalkanes of different length; no effect of the mutations was seen with dibromomethane, but an enhanced difference was observed with 1,3-dibromopropane and 1,5-dibromopentane. These results show that even slight alterations in the active site pocket of AGT that do not prevent its ability to protect cells from alkylating agents can block the paradoxical enhancement of the genotoxicity of the larger alpha,omega-dihaloalkanes by reducing the reaction with Cys145.


Subject(s)
Ethylene Dibromide/metabolism , Mutagens/toxicity , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Binding Sites , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Escherichia coli/drug effects , Escherichia coli/genetics , Half-Life , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Point Mutation , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...