Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Chem Res Toxicol ; 30(2): 669-677, 2017 02 20.
Article in English | MEDLINE | ID: mdl-27951635

ABSTRACT

DNA-protein cross-links are formed upon exposure of cellular DNA to various agents, including antitumor drugs, UV light, transition metals, and reactive oxygen species. They are thought to contribute to cancer, aging, and neurodegenerative diseases. It has been proposed that DNA-protein cross-links formed in cells are subject to proteolytic degradation to the corresponding DNA-peptide cross-links (DpCs). To investigate the effects of DpCs on DNA replication, we have constructed plasmid DNA containing a 10-mer Myc peptide covalently linked to C7 of 7-deaza-dG, a hydrolytically stable mimic of N7-dG lesions. Following transfection in human embryonic kidney cells (HEK 293T), progeny plasmids were recovered and sequenced. Translesion synthesis (TLS) past DpC was 76% compared to that of the unmodified control. The DpC induced 20% targeted G → A and G → T plus 15% semitargeted mutations, notably at a guanine (G5) five bases 3' to the lesion site. Proteolytic digestion of the DpC reduced the mutation frequency considerably, indicating that the covalently attached 10-mer peptide was responsible for the observed mutations. TLS efficiency and targeted mutations were reduced upon siRNA knockdown of pol η, pol κ, or pol ζ, indicating that they participate in error-prone bypass of the DpC lesion. However, the semitargeted mutation at G5 was only reduced upon knockdown of pol ζ, suggesting its critical role in this type of mutations. Our results indicate that DpCs formed at the N7 position of guanine can induce both targeted and semitargeted mutations in human cells and that the TLS polymerases play a critical role in their error-prone bypass.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase/metabolism , DNA/chemistry , Mutagens/chemistry , Peptides/chemistry , HEK293 Cells , Humans
2.
J Biol Chem ; 291(45): 23589-23603, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27621316

ABSTRACT

DNA-protein cross-links (DPCs) are bulky DNA lesions that form both endogenously and following exposure to bis-electrophiles such as common antitumor agents. The structural and biological consequences of DPCs have not been fully elucidated due to the complexity of these adducts. The most common site of DPC formation in DNA following treatment with bis-electrophiles such as nitrogen mustards and cisplatin is the N7 position of guanine, but the resulting conjugates are hydrolytically labile and thus are not suitable for structural and biological studies. In this report, hydrolytically stable structural mimics of N7-guanine-conjugated DPCs were generated by reductive amination reactions between the Lys and Arg side chains of proteins/peptides and aldehyde groups linked to 7-deazaguanine residues in DNA. These model DPCs were subjected to in vitro replication in the presence of human translesion synthesis DNA polymerases. DPCs containing full-length proteins (11-28 kDa) or a 23-mer peptide blocked human polymerases η and κ. DPC conjugates to a 10-mer peptide were bypassed with nucleotide insertion efficiency 50-100-fold lower than for native G. Both human polymerase (hPol) κ and hPol η inserted the correct base (C) opposite the 10-mer peptide cross-link, although small amounts of T were added by hPol η. Molecular dynamics simulation of an hPol κ ternary complex containing a template-primer DNA with dCTP opposite the 10-mer peptide DPC revealed that this bulky lesion can be accommodated in the polymerase active site by aligning with the major groove of the adducted DNA within the ternary complex of polymerase and dCTP.


Subject(s)
DNA Adducts/chemistry , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Guanine/analogs & derivatives , Peptides/chemistry , Proteins/chemistry , Amination , Amino Acid Sequence , Base Sequence , DNA Adducts/genetics , Guanine/chemistry , Humans , Molecular Dynamics Simulation , Oxidation-Reduction , Recombinant Proteins/metabolism
3.
Nucleic Acids Res ; 44(15): 7281-91, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27257072

ABSTRACT

Several important anti-tumor agents form DNA interstrand crosslinks (ICLs), but their clinical efficiency is counteracted by multiple complex DNA repair pathways. All of these pathways require unhooking of the ICL from one strand of a DNA duplex by nucleases, followed by bypass of the unhooked ICL by translesion synthesis (TLS) polymerases. The structures of the unhooked ICLs remain unknown, yet the position of incisions and processing of the unhooked ICLs significantly influence the efficiency and fidelity of bypass by TLS polymerases. We have synthesized a panel of model unhooked nitrogen mustard ICLs to systematically investigate how the state of an unhooked ICL affects pol η activity. We find that duplex distortion induced by a crosslink plays a crucial role in translesion synthesis, and length of the duplex surrounding an unhooked ICL critically affects polymerase efficiency. We report the synthesis of a putative ICL repair intermediate that mimics the complete processing of an unhooked ICL to a single crosslinked nucleotide, and find that it provides only a minimal obstacle for DNA polymerases. Our results raise the possibility that, depending on the structure and extent of processing of an ICL, its bypass may not absolutely require TLS polymerases.


Subject(s)
Cross-Linking Reagents/chemistry , DNA Damage , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , DNA/biosynthesis , DNA/chemistry , Nucleic Acid Conformation , DNA/metabolism , DNA Replication , DNA-Directed DNA Polymerase/genetics , Structure-Activity Relationship
4.
J Biol Chem ; 290(37): 22602-11, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26221031

ABSTRACT

Cisplatin and its derivatives, nitrogen mustards and mitomycin C, are used widely in cancer chemotherapy. Their efficacy is linked primarily to their ability to generate DNA interstrand cross-links (ICLs), which effectively block the progression of transcription and replication machineries. Release of this block, referred to as unhooking, has been postulated to require endonucleases that incise one strand of the duplex on either side of the ICL. Here we investigated how the 5' flap nucleases FANCD2-associated nuclease 1 (FAN1), exonuclease 1 (EXO1), and flap endonuclease 1 (FEN1) process a substrate reminiscent of a replication fork arrested at an ICL. We now show that EXO1 and FEN1 cleaved the substrate at the boundary between the single-stranded 5' flap and the duplex, whereas FAN1 incised it three to four nucleotides in the double-stranded region. This affected the outcome of processing of a substrate containing a nitrogen mustard-like ICL two nucleotides in the duplex region because FAN1, unlike EXO1 and FEN1, incised the substrate predominantly beyond the ICL and, therefore, failed to release the 5' flap. We also show that FAN1 was able to degrade a linear ICL substrate. This ability of FAN1 to traverse ICLs in DNA could help to elucidate its biological function, which is currently unknown.


Subject(s)
DNA Repair Enzymes/chemistry , DNA/chemistry , Exodeoxyribonucleases/chemistry , Flap Endonucleases/chemistry , Cell Line , DNA/genetics , DNA/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Endodeoxyribonucleases , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , Humans , Multifunctional Enzymes , Substrate Specificity
5.
Nucleic Acids Res ; 42(11): 7429-35, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24782532

ABSTRACT

DNA interstrand crosslinks (ICLs) are extremely cytotoxic lesions that block essential cellular processes, such as replication and transcription. Crosslinking agents are widely used in cancer chemotherapy and form an array of structurally diverse ICLs. Despite the clinical success of these agents, resistance of tumors to crosslinking agents, for example, through repair of these lesions by the cellular machinery remains a problem. We have previously reported the synthesis of site-specific ICLs mimicking those formed by nitrogen mustards to facilitate the studies of cellular responses to ICL formation. Here we extend these efforts and report the synthesis of structurally diverse major groove ICLs that induce severe, little or no distortion in the DNA. Our approach employs the incorporation of aldehyde precursors of different lengths into complementary strands and ICL formation using a double reductive amination with a variety of amines. Our studies provide insight into the structure and reactivity parameters of ICL formation by double reductive amination and yield a set of diverse ICLs that will be invaluable for exploring structure-activity relationships in ICL repair.


Subject(s)
Aldehydes/chemistry , DNA/chemistry , Chemistry Techniques, Synthetic , Mechlorethamine/chemistry , Models, Molecular , Nucleic Acid Conformation
6.
Mol Cell ; 54(3): 472-84, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24726326

ABSTRACT

SLX4 binds to three nucleases (XPF-ERCC1, MUS81-EME1, and SLX1), and its deficiency leads to genomic instability, sensitivity to DNA crosslinking agents, and Fanconi anemia. However, it is not understood how SLX4 and its associated nucleases act in DNA crosslink repair. Here, we uncover consequences of mouse Slx4 deficiency and reveal its function in DNA crosslink repair. Slx4-deficient mice develop epithelial cancers and have a contracted hematopoietic stem cell pool. The N-terminal domain of SLX4 (mini-SLX4) that only binds to XPF-ERCC1 is sufficient to confer resistance to DNA crosslinking agents. Recombinant mini-SLX4 enhances XPF-ERCC1 nuclease activity up to 100-fold, directing specificity toward DNA forks. Mini-SLX4-XPF-ERCC1 also vigorously stimulates dual incisions around a DNA crosslink embedded in a synthetic replication fork, an essential step in the repair of this lesion. These observations define vertebrate SLX4 as a tumor suppressor, which activates XPF-ERCC1 nuclease specificity in DNA crosslink repair.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Recombinases/physiology , Animals , Base Sequence , Bone Marrow Cells/pathology , DNA Adducts/chemistry , DNA Damage , DNA-Binding Proteins/chemistry , Endonucleases/chemistry , Hematopoietic Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/enzymology , Nucleic Acid Conformation , Tumor Suppressor Proteins
7.
Bioconjug Chem ; 24(9): 1496-506, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-23885807

ABSTRACT

DNA-protein cross-links (DPCs) are ubiquitous, structurally diverse DNA lesions formed upon exposure to bis-electrophiles, transition metals, UV light, and reactive oxygen species. Because of their superbulky, helix distorting nature, DPCs interfere with DNA replication, transcription, and repair, potentially contributing to mutagenesis and carcinogenesis. However, the biological implications of DPC lesions have not been fully elucidated due to the difficulty in generating site-specific DNA substrates representative of DPC lesions formed in vivo. In the present study, a novel approach involving postsynthetic reductive amination has been developed to prepare a range of hydrolytically stable lesions structurally mimicking the DPCs produced between the N7 position of guanine in DNA and basic lysine or arginine side chains of proteins and peptides.


Subject(s)
DNA/chemistry , Peptides/chemistry , Proteins/chemistry , Amination , Amino Acid Sequence , Animals , Humans , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...