Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 9(1): 24, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29295983

ABSTRACT

Tyrosyl-DNA phosphodiesterase (Tdp1) is a DNA 3'-end processing enzyme that repairs topoisomerase 1B-induced DNA damage. We use a new tool combining site-specific DNA-protein cross-linking with mass spectrometry to identify Tdp1 interactions with DNA. A conserved phenylalanine (F259) of Tdp1, required for efficient DNA processing in biochemical assays, cross-links to defined positions in DNA substrates. Crystal structures of Tdp1-DNA complexes capture the DNA repair machinery after 3'-end cleavage; these reveal how Tdp1 coordinates the 3'-phosphorylated product of nucleosidase activity and accommodates duplex DNA. A hydrophobic wedge splits the DNA ends, directing the scissile strand through a channel towards the active site. The F259 side-chain stacks against the -3 base pair, delimiting the junction of duplexed and melted DNA, and fixes the scissile strand in the channel. Our results explain why Tdp1 cleavage is non-processive and provide a molecular basis for DNA 3'-end processing by Tdp1.


Subject(s)
DNA Damage , DNA Repair , DNA/metabolism , Phosphoric Diester Hydrolases/metabolism , Base Sequence , Catalytic Domain , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , Humans , Models, Molecular , Nucleic Acid Conformation , Phosphoric Diester Hydrolases/chemistry , Protein Binding , Protein Domains
2.
Anal Chem ; 89(21): 11208-11213, 2017 11 07.
Article in English | MEDLINE | ID: mdl-28885003

ABSTRACT

Cross-linking of nucleic acids to proteins in combination with mass spectrometry permits the precise identification of interacting residues between nucleic acid-protein complexes. However, the mass spectrometric identification and characterization of cross-linked nucleic acid-protein heteroconjugates within a complex sample is challenging. Here we establish a novel enzymatic differential 16O/18O-labeling approach, which uniquely labels heteroconjugates. We have developed an automated data analysis workflow based on OpenMS for the identification of differentially isotopically labeled heteroconjugates against a complex background. We validated our method using synthetic model DNA oligonucleotide-peptide heteroconjugates, which were subjected to the labeling reaction and analyzed by high-resolution FTICR mass spectrometry.


Subject(s)
Fungal Proteins/chemistry , Mass Spectrometry/methods , Nucleoproteins/analysis , Single-Strand Specific DNA and RNA Endonucleases/chemistry , Trypsin/chemistry , Data Analysis , Isotope Labeling , Nucleoproteins/chemistry , Oxygen/chemistry , Oxygen Isotopes/chemistry , Software , Workflow
3.
Anal Chem ; 87(19): 9595-9, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26335278

ABSTRACT

UV cross-linking of nucleic acids to proteins in combination with mass spectrometry is a powerful technique to identify proteins, peptides, and the amino acids involved in intermolecular interactions within nucleic acid-protein complexes. However, the mass spectrometric identification of cross-linked nucleic acid-protein heteroconjugates in complex mixtures and MS/MS characterization of the specific sites of cross-linking is extremely challenging. As a tool for the optimization of sample preparation, ionization, fragmentation, and detection by mass spectrometry, novel synthetic DNA-peptide heteroconjugates were generated to act as mimics of UV cross-linked heteroconjugates. Click chemistry was employed to cross-link peptides to DNA oligonucleotides. These heteroconjugates were fully characterized by high resolution FTICR mass spectrometry and by collision-induced dissociation (CID) following nuclease P1 digestion of the DNA moiety to a single nucleotide monophosphate. This allowed the exact site of the cross-linking within the peptide to be unambiguously assigned. These synthetic DNA-peptide heteroconjugates have the potential to be of use for a variety of applications that involve DNA-peptide heteroconjugates.


Subject(s)
Click Chemistry , DNA/chemistry , Mass Spectrometry , Peptides/chemistry , Catalysis , Copper/chemistry , Molecular Structure
4.
J Biomol Screen ; 19(10): 1372-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25117203

ABSTRACT

Mutations of DNA repair pathways contribute to tumorigenesis and provide a therapeutic target for synthetic lethal interactions in tumor cells. Given that tyrosyl-DNA phosphodiesterase 1 (Tdp1) repairs stalled topoisomerase-I DNA complexes, we hypothesized that inhibition of Tdp1 has synthetic lethal effects in some cancers. To test this, we screened tumor arrays for Tdp1 expression and observed that Tdp1 is expressed in many tumors, including more than 90% of human breast tumors. Subsequent chemical screening identified putative Tdp1 inhibitors. Treatment of control human mammary epithelial cells and the breast cancer cell line MCF-7 with compound CD00509 preferentially sensitized MCF-7 cells to camptothecin and decreased cell proliferation 25% more than camptothecin treatment alone. This suggests that CD00509 specifically targeted Tdp1 in vitro, and CD00509 increased the sensitivity of wild-type murine embryonic fibroblasts (MEFs) to camptothecin to a degree comparable to that of Tdp1(-/-) MEFs. In addition, consistent with poly ADP-ribose polymerase-1 (PARP-1) collaborating with Tdp1 in DNA repair, combined Tdp1 and PARP-1 inhibition was more detrimental to MCF-7 cells than either treatment alone, whereas the combination was not additively harmful to control mammary cells. We conclude that targeting Tdp1 in anticancer therapy preferentially enhances the sensitivity of some breast cancer cells to camptothecin and may be an effective adjuvant for breast cancer therapy.


Subject(s)
High-Throughput Screening Assays/methods , Neoplasms/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pyrans/pharmacology , Thiobarbiturates/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Computer Simulation , DNA Damage/drug effects , Female , Gene Knockdown Techniques , Histones/metabolism , Humans , In Vitro Techniques , MCF-7 Cells/drug effects , Mice , Molecular Docking Simulation , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Tissue Array Analysis , Topoisomerase I Inhibitors/pharmacology
5.
Methods Mol Biol ; 1054: 173-85, 2013.
Article in English | MEDLINE | ID: mdl-23913292

ABSTRACT

Denaturing urea polyacrylamide gel electrophoresis (PAGE) allows the separation of linear single-stranded DNA molecules based on molecular weight. This method can be used to analyze or purify short synthesized DNA oligonucleotides or products from enzymatic reactions.In this chapter we describe how to prepare and how to run these high concentration polyacrylamide gels. We detail how to transfer a gel onto Whatman paper and how to dry it. Radiolabelled oligonucleotides are visualized by PhosphorImager technology.


Subject(s)
DNA/isolation & purification , Denaturing Gradient Gel Electrophoresis/methods , Oligonucleotides/isolation & purification , Electrophoresis, Polyacrylamide Gel , Urea/chemistry
6.
Biochem J ; 436(3): 559-66, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21463258

ABSTRACT

TDP (tyrosyl-DNA phosphodiesterase) 1 catalyses the hydrolysis of phosphodiester linkages between a DNA 3' phosphate and a tyrosine residue as well as a variety of other DNA 3' substituents, and has been implicated in the repair of covalent complexes involving eukaryotic type IB topoisomerases. To better understand the substrate features that are recognized by TDP1, the size of either the DNA or protein component of the substrate was varied. Competition experiments and gel-shift analyses comparing a series of substrates with DNA lengths increasing from 6 to 28 nt indicated that, contrary to predictions based on the crystal structure of the protein, the apparent affinity for the substrate increased as the DNA length was increased over the entire range tested. It has been found previously that a substrate containing the full-length native form of human topoisomerase I protein is not cleaved by TDP1. Protein-oligonucleotide complexes containing either a 53 or 108 amino acid topoisomerase I-derived peptide were efficiently cleaved by TDP1, but similar to the full-length protein, a substrate containing a 333 amino acid topoisomerase I fragment was resistant to cleavage. Consistent with these results, evidence is presented that processing by the proteasome is required for TDP1 cleavage in vivo.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Phosphoric Diester Hydrolases/metabolism , Humans , Peptide Fragments/metabolism , Substrate Specificity
7.
Adv Exp Med Biol ; 685: 75-83, 2010.
Article in English | MEDLINE | ID: mdl-20687496

ABSTRACT

Spinocerebellar ataxia with axonal neuropathy (SCAN 1) is an autosomal recessive disorder caused by a specific point mutation (c.1478A>G, p.H493R) in the tyrosyl-DNA phosphodiesterase (TDP1) gene. Functional and genetic studies suggest that this mutation, which disrupts the active site of the Tdp1 enzyme, causes disease by a combination of decreased catalytic activity and stabilization of the normally transient covalent Tdp1-DNA intermediate. This covalent reaction intermediate can form during the repair of stalled topoisomerase I-DNA adducts or oxidatively damaged bases at the 3' end of the DNA at a strand break. However, our current understanding of the biology of Tdp1 function in humans is limited and does not allow us to fully elucidate the disease mechanism.


Subject(s)
Chromosome Disorders/enzymology , DNA Repair-Deficiency Disorders/enzymology , Phosphoric Diester Hydrolases/metabolism , Point Mutation , Spinocerebellar Ataxias/enzymology , Chromosome Disorders/genetics , Chromosome Disorders/pathology , DNA/genetics , DNA/metabolism , DNA Breaks , DNA Repair/genetics , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/pathology , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Humans , Phosphoric Diester Hydrolases/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology
8.
J Med Chem ; 52(22): 7122-31, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19883083

ABSTRACT

Tyrosyl-DNA phosphodiesterase 1 (Tdp1) is an enzyme that catalyzes the hydrolysis of 3'-phosphotyrosyl bonds. Such linkages form in vivo when topoisomerase I (Top1) processes DNA. For this reason, Tdp1 has been implicated in the repair of irreversible Top1-DNA covalent complexes. Tdp1 inhibitors have been regarded as potential therapeutics in combination with Top1 inhibitors, such as the camptothecin derivatives, topotecan, and irinotecan, which are used to treat human cancers. Using a novel high-throughput screening assay, we have identified the C21-substituted progesterone derivative, NSC 88915 (1), as a potential Tdp1 inhibitor. Secondary screening and cross-reactivity studies with related DNA processing enzymes confirmed that compound 1 possesses specific Tdp1 inhibitory activity. Deconstruction of compound 1 into discrete functional groups reveals that both components are required for inhibition of Tdp1 activity. Moreover, the synthesis of analogues of compound 1 has provided insight into the structural requirements for the inhibition of Tdp1. Surface plasmon resonance shows that compound 1 binds to Tdp1, whereas an inactive analogue fails to interact with the enzyme. On the basis of molecular docking and mechanistic studies, we propose that these compounds are competitive inhibitors, which mimics the oligonucleotide-peptide Tdp1 substrate. These steroid derivatives represent a novel chemotype and provide a new scaffold for developing small molecule inhibitors of Tdp1.


Subject(s)
Benzenesulfonates/chemistry , Benzenesulfonates/pharmacology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pregnenediones/chemistry , Pregnenediones/pharmacology , Steroids/chemistry , Amino Acid Sequence , Animals , Axons/pathology , Benzenesulfonates/metabolism , Catalytic Domain , DNA, Single-Stranded/metabolism , Esters/chemistry , High-Throughput Screening Assays , Humans , Mice , Models, Molecular , Mutation , Phosphodiesterase Inhibitors/metabolism , Phosphoric Diester Hydrolases/chemistry , Pregnenediones/metabolism , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Surface Plasmon Resonance , Thermodynamics
9.
Proc Natl Acad Sci U S A ; 105(45): 17278-83, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18981420

ABSTRACT

Trypanosomatid RNA editing is a unique process and essential for these organisms. It therefore represents a drug target for a group of protozoa that includes the causative agents for African sleeping sickness and other devastating tropical and subtropical diseases. Here, we present drug-like inhibitors of a key enzyme in the editing machinery, RNA-editing ligase 1 (REL1). These inhibitors were identified through a strategy employing molecular dynamics to account for protein flexibility. A virtual screen of the REL1 crystal structure against the National Cancer Institute Diversity Set was performed by using AutoDock4. The top 30 compounds, predicted to interact with REL1's ATP-binding pocket, were further refined by using the relaxed complex scheme (RCS), which redocks the compounds to receptor structures extracted from an explicitly solvated molecular dynamics trajectory. The resulting reordering of the ligands and filtering based on drug-like properties resulted in an initial recommended set of 8 ligands, 2 of which exhibited micromolar activity against REL1. A subsequent hierarchical similarity search with the most active compound over the full National Cancer Institute database and RCS rescoring resulted in an additional set of 6 ligands, 2 of which were confirmed as REL1 inhibitors with IC(50) values of approximately 1 microM. Tests of the 3 most promising compounds against the most closely related bacteriophage T4 RNA ligase 2, as well as against human DNA ligase IIIbeta, indicated a considerable degree of selectivity for RNA ligases. These compounds are promising scaffolds for future drug design and discovery efforts against these important pathogens.


Subject(s)
Carbon-Oxygen Ligases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Ligands , Mitochondrial Proteins/antagonists & inhibitors , RNA Editing/genetics , Trypanosoma brucei brucei/metabolism , Animals , Carbon-Oxygen Ligases/chemistry , Computational Biology/methods , DNA Ligase ATP , DNA Ligases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Mitochondrial Proteins/chemistry , Molecular Structure , Poly-ADP-Ribose Binding Proteins , RNA Ligase (ATP)/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Xenopus Proteins
10.
EMBO J ; 26(22): 4732-43, 2007 Nov 14.
Article in English | MEDLINE | ID: mdl-17948061

ABSTRACT

Tyrosyl-DNA phosphodiesterase 1 (Tdp1) cleaves the phosphodiester bond between a covalently stalled topoisomerase I (Topo I) and the 3' end of DNA. Stalling of Topo I at DNA strand breaks is induced by endogenous DNA damage and the Topo I-specific anticancer drug camptothecin (CPT). The H493R mutation of Tdp1 causes the neurodegenerative disorder spinocerebellar ataxia with axonal neuropathy (SCAN1). Contrary to the hypothesis that SCAN1 arises from catalytically inactive Tdp1, Tdp1-/- mice are indistinguishable from wild-type mice, physically, histologically, behaviorally, and electrophysiologically. However, compared to wild-type mice, Tdp1-/- mice are hypersensitive to CPT and bleomycin but not to etoposide. Consistent with earlier in vitro studies, we show that the H493R Tdp1 mutant protein retains residual activity and becomes covalently trapped on the DNA after CPT treatment of SCAN1 cells. This result provides a direct demonstration that Tdp1 repairs Topo I covalent lesions in vivo and suggests that SCAN1 arises from the recessive neomorphic mutation H493R. This is a novel mechanism for disease since neomorphic mutations are generally dominant.


Subject(s)
Phosphoric Diester Hydrolases/metabolism , Spinocerebellar Ataxias/genetics , Animals , Antineoplastic Agents/pharmacology , Axons , Bleomycin/pharmacology , Brain/metabolism , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cells, Cultured , Comet Assay , Embryo, Mammalian/cytology , Etoposide/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Genes, Recessive , Humans , Irinotecan , Mice , Mice, Knockout , Mutation , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/genetics , Polyneuropathies/genetics , Polyneuropathies/metabolism , RNA, Messenger/metabolism , Spinocerebellar Ataxias/metabolism , Topotecan/pharmacology
11.
J Mol Biol ; 357(4): 1202-10, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16487540

ABSTRACT

Type IB topoisomerases are essential enzymes that are responsible for relaxing superhelical tension in DNA by forming a transient covalent nick in one strand of the DNA duplex. Topoisomerase I is a target for anti-cancer drugs such as camptothecin, and these drugs also target the topoisomerases I in pathogenic trypanosomes including Leishmania species and Trypanosoma brucei. Most eukaryotic enzymes, including human topoisomerase I, are monomeric. However, for Leishmania donovani, the DNA-binding activity and the majority of residues involved in catalysis are located in a large subunit, designated TOP1L, whereas the catalytic tyrosine residue responsible for covalent attachment to DNA is located in a smaller subunit, called TOP1S. Here, we present the 2.27A crystal structure of an active truncated L.donovani TOP1L/TOP1S heterodimer bound to nicked double-stranded DNA captured as a vanadate complex. The vanadate forms covalent linkages between the catalytic tyrosine residue of the small subunit and the nicked ends of the scissile DNA strand, mimicking the previously unseen transition state of the topoisomerase I catalytic cycle. This structure fills a critical gap in the existing ensemble of topoisomerase I structures and provides crucial insights into the catalytic mechanism.


Subject(s)
DNA Topoisomerases, Type I/chemistry , DNA/chemistry , Leishmania donovani/enzymology , Protein Structure, Quaternary , Protozoan Proteins/chemistry , Vanadates/chemistry , Animals , Arginine/chemistry , Catalytic Domain , Crystallography, X-Ray , DNA/metabolism , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Dimerization , Histidine/chemistry , Humans , Macromolecular Substances , Models, Molecular , Molecular Structure , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Topoisomerase I Inhibitors , Tyrosine/chemistry , Water/chemistry
12.
J Biol Chem ; 280(43): 36518-28, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16141202

ABSTRACT

Human tyrosyl-DNA phosphodiesterase (Tdp1) hydrolyzes the phosphodiester bond between a DNA 3' end and a tyrosyl moiety. In eukaryotic cells, this type of linkage is found in stalled topoisomerase I-DNA covalent complexes, and Tdp1 has been implicated in the repair of such complexes in vivo. We confirm here that the Tdp1 catalytic cycle involves a covalent reaction intermediate in which a histidine residue is connected to a DNA 3'-phosphate through a phosphoamide linkage. Most surprisingly, this linkage can be hydrolyzed by Tdp1, and unlike a topoisomerase I-DNA complex, which requires modification to be an efficient substrate for Tdp1, the native form of Tdp1 can be removed from the DNA. The spinocerebellar ataxia with axonal neuropathy neurodegenerative disease is caused by the H493R mutant form of Tdp1, which shows reduced enzymatic activity and accumulates the Tdp1-DNA covalent intermediate. The ability of wild type Tdp1 to remove the stalled mutant protein from the DNA likely explains the recessive nature of spinocerebellar ataxia with axonal neuropathy. In addition to its activity on phosphotyrosine and phosphohistidine substrates, Tdp1 also possesses a limited DNA and RNA 3'-exonuclease activity in which a single nucleoside is removed from the 3'-hydroxyl end of the substrate. Furthermore, Tdp1 also removes a 3' abasic site and an artificial 3'-biotin adduct from the DNA. In combination with earlier data showing that Tdp1 can use 3'-phosphoglycolate as a substrate, these data suggest that Tdp1 may function to remove a variety of 3' adducts from DNA during DNA repair.


Subject(s)
Phosphoamino Acids/chemistry , Phosphoric Diester Hydrolases/physiology , Alleles , Axons/pathology , Biotin/chemistry , Biotinylation , Catalysis , DNA/chemistry , DNA Repair , Furans/chemistry , Glycolates/chemistry , Humans , Hydrolysis , Models, Chemical , Models, Genetic , Mutation , Peptides/chemistry , Phosphoric Diester Hydrolases/metabolism , Protein Binding , RNA/chemistry , Substrate Specificity , Time Factors , Tyrosine/chemistry
13.
EMBO J ; 24(12): 2224-33, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15920477

ABSTRACT

Tyrosyl-DNA phosphodiesterase (Tdp1) catalyzes the hydrolysis of the tyrosyl-3' phosphate linkage found in topoisomerase I-DNA covalent complexes. The inherited disorder, spinocerebellar ataxia with axonal neuropathy (SCAN1), is caused by a H493R mutation in Tdp1. Contrary to earlier proposals that this disease results from a loss-of-function mutation, we show here that this mutation reduces enzyme activity approximately 25-fold and importantly causes the accumulation of the Tdp1-DNA covalent reaction intermediate. Thus, the attempted repair of topoisomerase I-DNA complexes by Tdp1 unexpectedly generates a new protein-DNA complex with an apparent half-life of approximately 13 min that, in addition to the unrepaired topoisomerase I-DNA complex, may interfere with transcription and replication in human cells and contribute to the SCAN1 phenotype. The analysis of Tdp1 mutant cell lines derived from SCAN1 patients reveals that they are hypersensitive to the topoisomerase I-specific anticancer drug camptothecin (CPT), implicating Tdp1 in the repair of CPT-induced topoisomerase I damage in human cells. This finding suggests that inhibitors of Tdp1 could act synergistically with CPT in anticancer therapy.


Subject(s)
Camptothecin/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoric Diester Hydrolases/genetics , Amino Acid Substitution , Flow Cytometry , Humans , Mutation , Neoplasms/drug therapy , Phosphoric Diester Hydrolases/metabolism , S Phase/drug effects
14.
J Biol Chem ; 279(53): 55618-25, 2004 Dec 31.
Article in English | MEDLINE | ID: mdl-15494395

ABSTRACT

Tyrosyl DNA phosphodiesterase 1 (TDP1) is a repair enzyme that removes adducts, e.g. of topoisomerase I from the 3'-phosphate of DNA breaks. When expressed in human cells as biofluorescent chimera, TDP1 appeared more mobile than topoisomerase I, less accumulated in nucleoli, and not chromosome-bound at early mitosis. Upon exposure to camptothecin both proteins were cleared from nucleoli and rendered less mobile in the nucleoplasm. However, with TDP1 this happened much more slowly reflecting most likely the redistribution of nucleolar structures upon inhibition of rDNA transcription. Thus, a steady association of TDP1 with topoisomerase I seems unlikely, whereas its integration into repair complexes assembled subsequently to the stabilization of DNA.topoisomerase I intermediates is supported. Cells expressing GFP-tagged TDP1 > 100-fold in excess of endogenous TDP1 exhibited a significant reduction of DNA damage induced by the topoisomerase I poison camptothecin and could be selected by that drug. Surprisingly, DNA damage induced by the topoisomerase II poison VP-16 was also diminished to a similar extent, whereas DNA damage independent of topoisomerase I or II was not affected. Overexpression of the inactive mutant GFP-TDP1(H263A) at similar levels did not reduce DNA damage by camptothecin or VP-16. These observations confirm a requirement of active TDP1 for the repair of topoisomerase I-mediated DNA damage. Our data also suggest a role of TDP1 in the repair of DNA damage mediated by topoisomerase II, which is less clear. Since overexpression of TDP1 did not compromise cell proliferation, it could be a pleiotropic resistance mechanism in cancer therapy.


Subject(s)
DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type I/metabolism , Phosphoric Diester Hydrolases/physiology , Bacterial Proteins/metabolism , Binding Sites , Blotting, Western , Camptothecin/chemistry , Camptothecin/pharmacology , Cell Line , Cell Line, Tumor , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Cloning, Molecular , DNA/metabolism , DNA Damage , DNA, Ribosomal/chemistry , Etoposide/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Luminescent Proteins/metabolism , Methylnitronitrosoguanidine/pharmacology , Microscopy, Fluorescence , Mitosis , Mutation , Phosphoric Diester Hydrolases/metabolism , Time Factors
15.
J Med Chem ; 47(4): 829-37, 2004 Feb 12.
Article in English | MEDLINE | ID: mdl-14761185

ABSTRACT

Tyrosyl-DNA phosphodiesterase (Tdp1) catalyzes the hydrolysis of a phosphodiester bond between a tyrosine residue and a DNA 3' phosphate and functions as a DNA repair enzyme that cleaves stalled topoisomerase I-DNA complexes. We previously determined a procedure to crystallize a quaternary complex containing Tdp1, vanadate, a DNA oligonucleotide, and a tyrosine-containing peptide that mimics the transition state for hydrolysis of the Tdp1 substrate. Here, the ability of vanadate to accept a variety of different ligands is exploited to produce several different quaternary complexes with a variety of oligonucleotides, and peptides or a tyrosine analogue, in efforts to explore the binding properties of the Tdp1 DNA and peptide binding clefts. Eight crystal structures of Tdp1 with vanadate, oligonucleotides, and peptides or peptide analogues were determined. These structures demonstrated that Tdp1 is able to bind substituents with limited sequence variation in the polypeptide moiety and also bind oligonucleotides with sequence variation at the 3' end. Additionally, the tyrosine analogue octopamine can replace topoisomerase I derived peptides as the apical ligand to vanadate. The versatility of this system suggests that the formation of quaternary complexes around vanadate could be adapted to become a useful method for structure-based inhibitor design and has the potential to be generally applicable to other enzymes that perform chemistry on phosphate esters.


Subject(s)
Oligonucleotides/chemistry , Peptides/chemistry , Phosphoric Diester Hydrolases/chemistry , Vanadates/chemistry , Binding Sites , Crystallography, X-Ray , DNA Topoisomerases, Type I/chemistry , Ligands , Models, Molecular , Octopamine/chemistry , Structure-Activity Relationship
16.
J Biol Chem ; 279(4): 2984-92, 2004 Jan 23.
Article in English | MEDLINE | ID: mdl-14594810

ABSTRACT

Based on co-crystal structures of human topoisomerase I with bound DNA, Lys(532) makes a minor groove contact with the strongly preferred thymidine residue at the site of covalent attachment (-1 position). Replacement of Lys(532) with either arginine or alanine has essentially no effect on the sequence preference of the enzyme, indicating that this interaction is not required for the preference for a T at the -1 position. Although both the cleavage and religation activities of the K532R mutant enzyme are reduced, cleavage is reduced to a greater extent than religation. The reverse is true for the K532A mutant enzyme with religation so impaired that the nicked intermediate accumulates during plasmid relaxation assays. Consistent with the shift in the cleavage religation equilibrium toward cleavage for the K532A mutant enzyme, expression of the mutant enzyme in Saccharomyces cerevisiae is cytotoxic, and thus this mutant enzyme mimics the effects of the anticancer drug camptothecin. Cleavage assays with the mutant enzymes using an oligonucleotide containing a 5'-bridging phosphorothiolate indicate that Lys(532) functions as a general acid during cleavage to protonate the leaving 5'-oxygen. It is possible that the contact with the -1 base is important during catalysis to provide positional rigidity to the active site. The corresponding residues in the vaccinia virus topoisomerase and the tyrosine recombinases may have similar critical roles in catalysis.


Subject(s)
DNA Topoisomerases, Type I/chemistry , Amino Acid Substitution , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Enzyme Activation , Humans , Hydrolysis , Lysine , Models, Molecular , Mutation , Protein Conformation , Saccharomyces cerevisiae , Substrate Specificity/genetics
17.
Chem Biol ; 10(2): 139-47, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12618186

ABSTRACT

Tyrosyl-DNA phosphodiesterase (Tdp1) is a member of the phospholipase D superfamily and acts as a DNA repair enzyme that removes stalled topoisomerase I- DNA complexes by hydrolyzing the bond between a tyrosine side chain and a DNA 3' phosphate. Despite the complexity of the substrate of this phosphodiesterase, vanadate succeeded in linking human Tdp1, a tyrosine-containing peptide, and a single-stranded DNA oligonucleotide into a quaternary complex that mimics the transition state for the first step of the catalytic reaction. The conformation of the bound substrate mimic gives compelling evidence that the topoisomerase I-DNA complex must undergo extensive modification prior to cleavage by Tdp1. The structure also illustrates that the use of vanadate as the central moiety in high-order complexes has the potential to be a general method for capturing protein-substrate interactions for phosphoryl transfer enzymes, even when the substrates are large, complicated, and unusual.


Subject(s)
DNA Topoisomerases, Type I/chemistry , DNA/metabolism , Phosphoric Diester Hydrolases/chemistry , Vanadates/chemistry , Crystallization , Crystallography, X-Ray , Humans , Hydrogen Bonding , Models, Molecular , Molecular Mimicry , Oligonucleotides/chemistry , Protein Conformation
18.
J Mol Biol ; 324(5): 917-32, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12470949

ABSTRACT

Tyrosyl-DNA phosphodiesterase (Tdp1) is a DNA repair enzyme that catalyzes the hydrolysis of a phosphodiester bond between a tyrosine residue and a DNA 3'-phosphate. The only known example of such a linkage in eukaryotic cells occurs normally as a transient link between a type IB topoisomerase and DNA. Thus human Tdp1 is thought to be responsible for repairing lesions that occur when topoisomerase I becomes stalled on the DNA in the cell. Tdp1 has also been shown to remove glycolate from single-stranded DNA containing a 3'-phosphoglycolate, suggesting a role for Tdp1 in repair of free-radical mediated DNA double-strand breaks. We report the three-dimensional structures of human Tdp1 bound to the phosphate transition state analogs vanadate and tungstate. Each structure shows the inhibitor covalently bound to His263, confirming that this residue is the nucleophile in the first step of the catalytic reaction. Vanadate in the Tdp1-vanadate structure has a trigonal bipyramidal geometry that mimics the transition state for hydrolysis of a phosphodiester bond, while Tdp1-tungstate displays unusual octahedral coordination. The presence of low-occupancy tungstate molecules along the narrow groove of the substrate binding cleft is suggestive evidence that this groove binds ssDNA. In both cases, glycerol from the cryoprotectant solution became liganded to the vanadate or tungstate inhibitor molecules in a bidentate 1,2-diol fashion. These structural models allow predictions to be made regarding the specific binding mode of the substrate and the mechanism of catalysis.


Subject(s)
Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Tungsten Compounds/metabolism , Tungsten Compounds/pharmacology , Vanadates/metabolism , Vanadates/pharmacology , Apoenzymes/antagonists & inhibitors , Apoenzymes/chemistry , Apoenzymes/metabolism , Binding Sites , Catalysis/drug effects , Dose-Response Relationship, Drug , Humans , Hydrogen Bonding , Models, Chemical , Models, Molecular , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/pharmacology , Protein Conformation , Substrate Specificity , Tungsten Compounds/chemistry , Vanadates/chemistry
19.
Structure ; 10(2): 237-48, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839309

ABSTRACT

Tyrosyl-DNA phosphodiesterase (Tdp1) catalyzes the hydrolysis of a phosphodiester bond between a tyrosine residue and a DNA 3' phosphate. The enzyme appears to be responsible for repairing the unique protein-DNA linkage that occurs when eukaryotic topoisomerase I becomes stalled on the DNA in the cell. The 1.69 A crystal structure reveals that human Tdp1 is a monomer composed of two similar domains that are related by a pseudo-2-fold axis of symmetry. Each domain contributes conserved histidine, lysine, and asparagine residues to form a single active site. The structure of Tdp1 confirms that the protein has many similarities to the members of the phospholipase D (PLD) superfamily and indicates a similar catalytic mechanism. The structure also suggests how the unusual protein-DNA substrate binds and provides insights about the nature of the substrate in vivo.


Subject(s)
Phosphoric Diester Hydrolases/chemistry , Amino Acid Sequence , Bacterial Proteins/chemistry , Binding Sites , Catalysis , Crystallography, X-Ray , Humans , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Phospholipase D/chemistry , Phosphoric Diester Hydrolases/metabolism , Protein Conformation , Sequence Homology, Amino Acid , Static Electricity , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...