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










Publication year range
1.
J Transl Med ; 21(1): 738, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37858134

ABSTRACT

BACKGROUND: Autophagy is involved in nasopharyngeal carcinoma (NPC) radioresistance. Replication protein A 1 (RPA1) and RPA3, substrates of the RPA complex, are potential therapeutic targets for reversing NPC radioresistance. Nevertheless, the role of RPA in autophagy is not adequately understood. This investigation was performed to reveal the cytotoxic mechanism of a pharmacologic RPA inhibitor (RPAi) in NPC cells and the underlying mechanism by which RPAi-mediated autophagy regulates NPC radiosensitivity. METHODS AND RESULTS: We characterized a potent RPAi (HAMNO) that was substantially correlated with radiosensitivity enhancement and proliferative inhibition of in vivo and in NPC cell lines in vitro. We show that the RPAi induced autophagy at multiple levels by inducing autophagic flux, AMPK/mTOR pathway activation, and autophagy-related gene transcription by decreasing glycolytic function. We hypothesized that RPA inhibition impaired glycolysis and increased NPC dependence on autophagy. We further demonstrated that combining autophagy inhibition with chloroquine (CQ) treatment or genetic inhibition of the autophagy regulator ATG5 and RPAi treatment was more effective than either approach alone in enhancing the antitumor response of NPC to radiation. CONCLUSIONS: Our study suggests that HAMNO is a potent RPAi that enhances radiosensitivity and induces autophagy in NPC cell lines by decreasing glycolytic function and activating autophagy-related genes. We suggest a novel treatment strategy in which pharmacological inhibitors that simultaneously disrupt RPA and autophagic processes improve NPC responsiveness to radiation.


Subject(s)
Antineoplastic Agents , Autophagy , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Radiation Tolerance , Replication Protein A , Humans , Antineoplastic Agents/therapeutic use , Apoptosis , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Replication Protein A/antagonists & inhibitors , Replication Protein A/genetics , Autophagy-Related Protein 5/genetics
2.
Methods Mol Biol ; 1999: 217-221, 2019.
Article in English | MEDLINE | ID: mdl-31127579

ABSTRACT

With the recent interest in targeting the DNA damage response (DDR) and DNA repair, new screening methodologies are needed to broaden the scope of targetable proteins beyond kinases and traditional enzymes. Many of the proteins involved in the DDR and repair impart their activity by making specific contacts with DNA. These protein-nucleic acid interactions represent a tractable target for perturbation with small molecules. We describe a high throughput, solution-based equilibrium binding fluorescence polarization assay that can be applied to a wide array of protein-nucleic acid interactions. The assay is sensitive, stable, and able to identify small molecules capable of blocking DNA-protein interactions.


Subject(s)
DNA Repair/drug effects , High-Throughput Screening Assays/methods , Replication Protein A/antagonists & inhibitors , Xeroderma Pigmentosum Group A Protein/antagonists & inhibitors , DNA/genetics , DNA/metabolism , DNA Damage , Fluorescence Polarization/methods , Protein Binding/drug effects , Protein Binding/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
3.
J Mol Graph Model ; 86: 149-159, 2019 01.
Article in English | MEDLINE | ID: mdl-30366191

ABSTRACT

Replication Protein A (RPA) mediates DNA Damage Response (DDR) pathways through protein-protein interactions (PPIs). Targeting the PPIs formed between RPA and other DNA Damage Response (DDR) mediators has become an intriguing area of research for cancer drug discovery. A number of studies applied different methods ranging from high throughput screening approaches to fragment-based drug design tools to discover RPA inhibitors. Although these methods are robust, virtual screening approaches may be allocated as an alternative to such experimental methods, especially for screening of large libraries. Here we report the comprehensive screening of the large database, ZINC15 composed of ∼750 M compounds and the comparison of the identified ligands with the previously known inhibitors by means of binding affinity and drug-likeness. Initially, a ligand library sharing similarity with a promising inhibitor of the N-terminal domain of the RPA70 subunit (RPA70N) was generated by screening of the ZINC15 library. 46,999 ligands were collected and screened by LeDock which produced a satisfactory correlation with the experimental values (R2 = 0.77). 10 of the top-scoring ligands in LeDock were directly progressed to molecular dynamics (MD) simulations, while 10 additional ligands were also selected based on their LeDock scores and the presence of a functional group that could interact with the key amino acids in the RPA70N cleft. MD simulations were used to predict the binding free energy of the ligands by the MM-PBSA method which produced a high level of agreement with the experiments (R2 = 0.85). Binding free energy predictions pointed out 2 ligands with higher binding affinity than any of the reference inhibitors. Particularly the ligand ZINC000753854163 exhibited superior drug-likeness features than any of the known inhibitors. Overall, this study reports ZINC000753854163 as a possible inhibitor of RPA70N, reflecting its possible use in RPA70N targeted cancer therapy.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Interaction Domains and Motifs , Replication Protein A/chemistry , Binding Sites , Drug Discovery/methods , Humans , Ligands , Protein Binding , Protein Interaction Domains and Motifs/drug effects , Quantitative Structure-Activity Relationship , Replication Protein A/antagonists & inhibitors , Small Molecule Libraries
4.
Exp Cell Res ; 371(2): 330-341, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30144445

ABSTRACT

BACKGROUND/AIM: Nasopharyngeal cancer (NPC) has a high local recurrence rate due to its resistance to ionizing radiation (IR). Replication protein A1 (RPA1) is one of the main elements in the homologous repair (HR) pathway, which is closely associated with the repair of DNA double strand breaks (DDBs). Studies on the relationship between RPA1 and the radiosensitivity of NPC are substantially limited. It was hypothesized that RPA1 plays a crucial role in predicting the radiosensitivity of NPC. METHODS: The protein expression of RPA1 in 182 patients with NPC in the complete response (CR) and non-complete response (nCR) groups was evaluated using immunohistochemistry. Then, univariate and multivariate analysis were performed using SPSS software vision 22 to determine the relationship between the expression of RPA1 and the clinicopathological features. In addition, the mRNA expression of RPA1 was tested in 24 fresh samples using qRT-PCR. RPA1 was silenced in CNE-2R cell lines combined with IR to measure the radiosensitivity, proliferation, DNA damage repair and cell cycle of CNE-2R cells. Xenograft models in nude mice were used to determine the effect of RPA1 on tumor growth after IR. Immunoblotting and immunofluorescence staining were performed to identify proteins that interacted with RPA1. All statistical tests were two-sided. RESULTS: RPA1 protein was overexpressed in NPC patients with nCR (65.31%), and was an independent predictor of radiosensitivity (HR: 3.755, 95% CI: 1.990-7.085), in addition to Epstein-Barr virus (EBV; HR: 3.984; 95% CI: 1.524-10.410). The silencing of RPA1 increased the radiosensitivity of CNE-2R cells, blocked the repair of DNA, impaired cell proliferation, and contributed to G2/M cell cycle arrest. Furthermore, the xenograft models in nude mice revealed that silencing RPA1 combined with irradiation significantly retarded the growth of tumors. Moreover, the knockdown of RPA1 decreased Rad51 collection to the damage site and prolonged the time of DNA repair. CONCLUSION: RPA1 protein is frequently overexpressed in NPC patients with nCR. The silencing of RPA1 enhanced the radiosensitivity of CNE-2R cells. These present findings reveal that RPA1 is a potential biomarker for predicting the radiosensitivity in NPC.


Subject(s)
Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Rad51 Recombinase/genetics , Radiation Tolerance/genetics , Replication Protein A/genetics , Adult , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cisplatin/therapeutic use , DNA Damage , Female , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/radiation effects , Humans , Male , Mice , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/surgery , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/surgery , Nasopharyngeal Neoplasms/therapy , Neoplasm Staging , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism , Radiation Tolerance/drug effects , Replication Protein A/antagonists & inhibitors , Replication Protein A/metabolism , Retrospective Studies , Signal Transduction , Tumor Burden/drug effects , Tumor Burden/radiation effects , X-Rays , Xenograft Model Antitumor Assays
5.
Nat Struct Mol Biol ; 24(12): 1116-1123, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058711

ABSTRACT

Mammalian polymerase theta (Polθ) is a multifunctional enzyme that promotes error-prone DNA repair by alternative nonhomologous end joining (alt-NHEJ). Here we present structure-function analyses that reveal that, in addition to the polymerase domain, Polθ-helicase activity plays a central role during double-strand break (DSB) repair. Our results show that the helicase domain promotes chromosomal translocations by alt-NHEJ in mouse embryonic stem cells and also suppresses CRISPR-Cas9- mediated gene targeting by homologous recombination (HR). In vitro assays demonstrate that Polθ-helicase activity facilitates the removal of RPA from resected DSBs to allow their annealing and subsequent joining by alt-NHEJ. Consistent with an antagonistic role for RPA during alt-NHEJ, inhibition of RPA1 enhances end joining and suppresses recombination. Taken together, our results reveal that the balance between HR and alt-NHEJ is controlled by opposing activities of Polθ and RPA, providing further insight into the regulation of repair-pathway choice in mammalian cells.


Subject(s)
Catalytic Domain/genetics , DNA End-Joining Repair/genetics , DNA-Directed DNA Polymerase/genetics , Embryonic Stem Cells/cytology , Replication Protein A/antagonists & inhibitors , Animals , CRISPR-Cas Systems/genetics , Cell Line , DNA Breaks, Double-Stranded , Homologous Recombination/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Replication Protein A/genetics , Structure-Activity Relationship , Translocation, Genetic/genetics , DNA Polymerase theta
6.
DNA Repair (Amst) ; 58: 1-12, 2017 10.
Article in English | MEDLINE | ID: mdl-28837865

ABSTRACT

It was reported that valproic acid (VPA, a histone deacetylase inhibitor) can sensitize cancer cells to hydroxyurea (HU, a ribonucleotide reductase inhibitor) for chemotherapy, although the mechanism of VPA-induced HU sensitization is unclear. In this study, we systematically characterized VPA-induced HU sensitization of breast cancer cells. Multiple breast cancer cell models were employed to investigate whether the safe concentration of 0.5mM VPA and 2mM HU can result in DNA double-strand breaks (DSBs) and impact cell survival. Furthermore, the underlying mechanism was explored through cell biology assays, including clonogenic survival, homologous recombination (HR) activity, immunoblot and immunofluorescence. We found that VPA and HU cooperatively suppressed cancer cell survival. VPA resulted in the accumulation of more DNA double-strand breaks (DSBs) in response to HU-induced replication arrest and was able to block HU-stimulated homologous recombination (HR) through inhibiting the activity of two key HR repair proteins by hyperphosphorylation of replication protein A2 (RPA2-p) and recombinase Rad51. However, apoptosis was not detected under this condition. In addition, the results from the survival fraction in the cells expressing defective RPA2-p showed that VPA disrupted the HU-induced RPA2-p-Rad51-mediated HR pathway. Importantly, these findings were further supported by analyzing primary-culture cells from the tissue of chemical carcinogen (DMBA)-induced breast cancer in rats. Thus, our data demonstrated that VPA and HU synergistically suppressed tumor cells via disturbing RPA2-p-mediated DNA repair pathway, which provides a new way for combining chemotherapeutic drugs to sensitize breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Hydroxyurea/therapeutic use , Recombinational DNA Repair/drug effects , Replication Protein A/antagonists & inhibitors , Valproic Acid/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Replication/drug effects , Female , Humans , Rad51 Recombinase/metabolism , Rats , Replication Protein A/metabolism , Valproic Acid/pharmacology
7.
FEBS Lett ; 591(8): 1083-1100, 2017 04.
Article in English | MEDLINE | ID: mdl-28079255

ABSTRACT

Coordination between DNA replication and DNA repair ensures maintenance of genome integrity, which is lost in cancer cells. Emerging evidence has linked homologous recombination (HR) proteins RAD51, BRCA1 and BRCA2 to the stability of nascent DNA. This function appears to be distinct from double-strand break (DSB) repair and is in part due to the prevention of MRE11-mediated degradation of nascent DNA at stalled forks. The role of RAD51 in fork protection resembles the activity described for its prokaryotic orthologue RecA, which prevents nuclease-mediated degradation of DNA and promotes replication fork restart in cells challenged by DNA-damaging agents. Here, we examine the mechanistic aspects of HR-mediated fork protection, addressing the crosstalk between HR and replication proteins.


Subject(s)
BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , DNA Replication , DNA-Binding Proteins/antagonists & inhibitors , Homologous Recombination , Models, Biological , Rad51 Recombinase/metabolism , Acid Anhydride Hydrolases , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromosomal Instability , DNA Breaks , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , MRE11 Homologue Protein , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Multimerization , Replication Protein A/antagonists & inhibitors , Replication Protein A/chemistry , Replication Protein A/metabolism
8.
Nucleic Acids Res ; 44(12): 5743-57, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27131361

ABSTRACT

DNA double-strand breaks (DSBs) are toxic lesions, which if improperly repaired can result in cell death or genomic instability. DSB repair is usually facilitated by the classical non-homologous end joining (C-NHEJ), or homologous recombination (HR) pathways. However, a mutagenic alternative NHEJ pathway, microhomology-mediated end joining (MMEJ), can also be deployed. While MMEJ is suppressed by C-NHEJ, the relationship between HR and MMEJ is less clear. Here, we describe a role for HR genes in suppressing MMEJ in human cells. By monitoring DSB mis-repair using a sensitive HPRT assay, we found that depletion of HR proteins, including BRCA2, BRCA1 or RPA, resulted in a distinct mutational signature associated with significant increases in break-induced mutation frequencies, deletion lengths and the annealing of short regions of microhomology (2-6 bp) across the break-site. This signature was dependent on CtIP, MRE11, POLQ and PARP, and thus indicative of MMEJ. In contrast to CtIP or MRE11, depletion of BRCA1 resulted in increased partial resection and MMEJ, thus revealing a functional distinction between these early acting HR factors. Together these findings indicate that HR factors suppress mutagenic MMEJ following DSB resection.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA/metabolism , Recombinational DNA Repair , Replication Protein A/genetics , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/metabolism , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/metabolism , Base Sequence , Biological Assay , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Endodeoxyribonucleases , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , MRE11 Homologue Protein , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Replication Protein A/antagonists & inhibitors , Replication Protein A/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , DNA Polymerase theta
9.
ChemMedChem ; 11(8): 893-9, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26748787

ABSTRACT

Replication protein A (RPA) is an essential single-stranded DNA (ssDNA)-binding protein that initiates the DNA damage response pathway through protein-protein interactions (PPIs) mediated by its 70N domain. The identification and use of chemical probes that can specifically disrupt these interactions is important for validating RPA as a cancer target. A high-throughput screen (HTS) to identify new chemical entities was conducted, and 90 hit compounds were identified. From these initial hits, an anthranilic acid based series was optimized by using a structure-guided iterative medicinal chemistry approach to yield a cell-penetrant compound that binds to RPA70N with an affinity of 812 nm. This compound, 2-(3- (N-(3,4-dichlorophenyl)sulfamoyl)-4-methylbenzamido)benzoic acid (20 c), is capable of inhibiting PPIs mediated by this domain.


Subject(s)
Replication Protein A/antagonists & inhibitors , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/pharmacology , Anisotropy , Dose-Response Relationship, Drug , Fluorescence Polarization , High-Throughput Screening Assays , Models, Molecular , Molecular Structure , Structure-Activity Relationship , ortho-Aminobenzoates/chemical synthesis
10.
Cell Signal ; 28(3): 214-223, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26691982

ABSTRACT

The RecQ helicases play roles in maintenance of genomic stability in species ranging from Escherichia coli to humans and interact with proteins involved in DNA metabolic pathways such as DNA repair, recombination, and replication. Our previous studies found that the Caenorhabditis elegans WRN-1 RecQ protein (a human WRN ortholog) exhibits ATP-dependent 3'-5' helicase activity and that the WRN-1 helicase is stimulated by RPA-1 on a long forked DNA duplex. However, the role of WRN-1 in response to S-phase associated with DSBs is unclear. We found that WRN-1 is involved in the checkpoint response to DSBs after CPT, inducing cell cycle arrest, is recruited to DSBs by RPA-1 and functions upstream of ATL-1 and ATM-1 for CHK-1 phosphorylation in the S-phase checkpoint. In addition, WRN-1 and RPA-1 recruitments to the DSBs require MRE-11, suggesting that DSB processing controlled by MRE-11 is important for WRN-1 at DSBs. The repair of CPT-induced DSBs is greatly reduced in the absence of WRN-1. These observations suggest that WRN-1 functions downstream of RPA-1 and upstream of CHK-1 in the DSB checkpoint pathway and is also required for the repair of DSB.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Camptothecin/toxicity , DNA Breaks, Double-Stranded/drug effects , DNA Helicases/metabolism , DNA Repair , Animals , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Checkpoint Kinase 1 , Comet Assay , DNA Helicases/genetics , Mutagenesis , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Interference , Replication Protein A/antagonists & inhibitors , Replication Protein A/genetics , Replication Protein A/metabolism , S Phase Cell Cycle Checkpoints/drug effects
11.
Nucleic Acids Res ; 43(10): 4962-74, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25916848

ABSTRACT

The primary eukaryotic single-stranded DNA-binding protein, Replication protein A (RPA), binds to single-stranded DNA at the sites of DNA damage and recruits the apical checkpoint kinase, ATR via its partner protein, ATRIP. It has been demonstrated that absence of RPA incapacitates the ATR-mediated checkpoint response. We report that in the absence of RPA, human single-stranded DNA-binding protein 1 (hSSB1) and its partner protein INTS3 form sub-nuclear foci, associate with the ATR-ATRIP complex and recruit it to the sites of genomic stress. The ATRIP foci formed after RPA depletion are abrogated in the absence of INTS3, establishing that hSSB-INTS3 complex recruits the ATR-ATRIP checkpoint complex to the sites of genomic stress. Depletion of homologs hSSB1/2 and INTS3 in RPA-deficient cells attenuates Chk1 phosphorylation, indicating that the cells are debilitated in responding to stress. We have identified that TopBP1 and the Rad9-Rad1-Hus1 complex are essential for the alternate mode of ATR activation. In summation, we report that the single-stranded DNA-binding protein complex, hSSB1/2-INTS3 can recruit the checkpoint complex to initiate ATR signaling.


Subject(s)
DNA-Binding Proteins/metabolism , Mitochondrial Proteins/metabolism , Replication Protein A/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Checkpoint Kinase 1 , DNA, Single-Stranded/metabolism , HeLa Cells , Humans , Protein Kinases/metabolism , RNA Interference , Replication Protein A/genetics , Signal Transduction , Stress, Physiological/genetics
12.
Cancer Res ; 74(18): 5165-72, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25070753

ABSTRACT

The ATR/Chk1 pathway is a critical surveillance network that maintains genomic integrity during DNA replication by stabilizing the replication forks during normal replication to avoid replication stress. One of the many differences between normal cells and cancer cells is the amount of replication stress that occurs during replication. Cancer cells with activated oncogenes generate increased levels of replication stress. This creates an increased dependency on the ATR/Chk1 pathway in cancer cells and opens up an opportunity to preferentially kill cancer cells by inhibiting this pathway. In support of this idea, we have identified a small molecule termed HAMNO ((1Z)-1-[(2-hydroxyanilino)methylidene]naphthalen-2-one), a novel protein interaction inhibitor of replication protein A (RPA), a protein involved in the ATR/Chk1 pathway. HAMNO selectively binds the N-terminal domain of RPA70, effectively inhibiting critical RPA protein interactions that rely on this domain. HAMNO inhibits both ATR autophosphorylation and phosphorylation of RPA32 Ser33 by ATR. By itself, HAMNO treatment creates DNA replication stress in cancer cells that are already experiencing replication stress, but not in normal cells, and it acts synergistically with etoposide to kill cancer cells in vitro and slow tumor growth in vivo. Thus, HAMNO illustrates how RPA inhibitors represent candidate therapeutics for cancer treatment, providing disease selectivity in cancer cells by targeting their differential response to replication stress. Cancer Res; 74(18); 5165-72. ©2014 AACR.


Subject(s)
DNA Replication/drug effects , Naphthalenes/pharmacology , Replication Protein A/antagonists & inhibitors , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Growth Processes/drug effects , Checkpoint Kinase 1 , DNA Damage , Female , Humans , Mice , Mice, Nude , Models, Molecular , Phosphorylation , Protein Kinases/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Signal Transduction/drug effects
13.
Biochemistry ; 52(37): 6515-24, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23962067

ABSTRACT

Replication protein A (RPA) is the primary single-stranded DNA (ssDNA) binding protein in eukaryotes. The N-terminal domain of the RPA70 subunit (RPA70N) interacts via a basic cleft with a wide range of DNA processing proteins, including several that regulate DNA damage response and repair. Small molecule inhibitors that disrupt these protein-protein interactions are therefore of interest as chemical probes of these critical DNA processing pathways and as inhibitors to counter the upregulation of DNA damage response and repair associated with treatment of cancer patients with radiation or DNA-damaging agents. Determination of three-dimensional structures of protein-ligand complexes is an important step for elaboration of small molecule inhibitors. However, although crystal structures of free RPA70N and an RPA70N-peptide fusion construct have been reported, RPA70N-inhibitor complexes have been recalcitrant to crystallization. Analysis of the P61 lattice of RPA70N crystals led us to hypothesize that the ligand-binding surface was occluded. Surface reengineering to alter key crystal lattice contacts led to the design of RPA70N E7R, E100R, and E7R/E100R mutants. These mutants crystallized in a P212121 lattice that clearly had significant solvent channels open to the critical basic cleft. Analysis of X-ray crystal structures, target peptide binding affinities, and (15)N-(1)H heteronuclear single-quantum coherence nuclear magnetic resonance spectra showed that the mutations do not result in perturbations of the RPA70N ligand-binding surface. The success of the design was demonstrated by determining the structure of RPA70N E7R soaked with a ligand discovered in a previously reported molecular fragment screen. A fluorescence anisotropy competition binding assay revealed this compound can inhibit the interaction of RPA70N with the peptide binding motif from the DNA damage response protein ATRIP. The implications of the results are discussed in the context of ongoing efforts to design RPA70N inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Replication Protein A/genetics , Adaptor Proteins, Signal Transducing/chemistry , Crystallization , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Fluorescence Polarization , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs , Replication Protein A/antagonists & inhibitors , Replication Protein A/chemistry , Static Electricity
14.
PLoS One ; 7(7): e41208, 2012.
Article in English | MEDLINE | ID: mdl-22911760

ABSTRACT

RPA is a single-stranded DNA binding protein that physically associates with the BLM complex. RPA stimulates BLM helicase activity as well as the double Holliday junction dissolution activity of the BLM-topoisomerase IIIα complex. We investigated the effect of RPA on the ssDNA decatenase activity of topoisomerase IIIα. We found that RPA and other ssDNA binding proteins inhibit decatenation by topoisomerase IIIα. Complex formation between BLM, TopoIIIα, and RMI1 ablates inhibition of decatenation by ssDNA binding proteins. Together, these data indicate that inhibition by RPA does not involve species-specific interactions between RPA and BLM-TopoIIIα-RMI1, which contrasts with RPA modulation of double Holliday junction dissolution. We propose that topoisomerase IIIα and RPA compete to bind to single-stranded regions of catenanes. Interactions with BLM and RMI1 enhance toposiomerase IIIα activity, promoting decatenation in the presence of RPA.


Subject(s)
Carrier Proteins/metabolism , DNA Topoisomerases, Type I/metabolism , RecQ Helicases/metabolism , Replication Protein A/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , Escherichia coli Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Binding , Replication Protein A/antagonists & inhibitors
15.
Anal Biochem ; 421(2): 742-9, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22197419

ABSTRACT

Replication protein A (RPA) interacts with multiple checkpoint proteins and promotes signaling through the ATR kinase, a key regulator of checkpoint pathways in the mammalian response to DNA damage. In cancer cells, increased DNA repair activity contributes to resistance to chemotherapy. Therefore, small molecules that block binding of checkpoint proteins to RPA may inhibit the DNA damage response and, thus, sensitize cancer cells to DNA-damaging agents. Here we report on the development of a homogeneous, high-throughput fluorescence polarization assay for identifying compounds that block the critical protein-protein interaction site in the basic cleft of the 70N domain of RPA (RPA70N). A fluorescein isothiocyanate (FITC)-labeled peptide derived from the ATR cofactor, ATRIP, was used as a probe in the binding assay. The ability of the assay to accurately detect relevant ligands was confirmed using peptides derived from ATRIP, RAD9, MRE11, and p53. The assay was validated for use in high-throughput screening using the Spectrum collection of 2000 compounds. The FPA assay was performed with a Z' factor of ≥ 0.76 in a 384-well format and identified several compounds capable of inhibiting the RPA70N binding interface.


Subject(s)
Fluorescence Polarization/methods , High-Throughput Screening Assays , Replication Protein A/chemistry , Amino Acid Sequence , DNA Damage , Molecular Sequence Data , Protein Structure, Tertiary , Replication Protein A/antagonists & inhibitors
16.
Mol Cancer Ther ; 10(10): 1796-806, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21846830

ABSTRACT

Replication protein A (RPA) is a single-strand DNA-binding protein with essential roles in DNA replication, recombination, and repair. It is necessary for the formation of the preincision complex that is required for proper incision of damaged DNA nucleotides during DNA repair. We have previously identified small molecule inhibitors (SMI) with the ability to disrupt RPA-binding activity to ssDNA. Further characterization of these RPA inhibitors was done using both lung and ovarian cancer cell lines. Lung cancer cell lines showed increased apoptotic cell death following treatment with the SMI MCI13E, with IC(50) values of approximately 5 µmol/L. The ovarian cancer cell line A2780 and the p53-null lung cancer cell line H1299 were particularly sensitive to MCI13E treatment, with IC(50) values less than 3 µmol/L. Furthermore, a cell-cycle effect was observed in lung cancer cell lines that resulted in a lengthening of either G(1) or S-phases of the cell cycle following single-agent treatment. Sequential treatment with MCI13E and cisplatin resulted in synergism. Overall, these data suggest that decreasing DNA-binding activity of RPA via a SMI may disrupt the role of RPA in cell-cycle regulation. Thus, SMIs of RPA hold the potential to be used as single-agent chemotherapeutics or in combination with current chemotherapeutic regimens to increase efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camphanes/chemistry , Camphanes/pharmacology , Cisplatin/pharmacology , DNA Replication/drug effects , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Replication Protein A/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Damage , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Drug Synergism , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Replication Protein A/chemistry , Replication Protein A/genetics , Replication Protein A/metabolism , Small Molecule Libraries
17.
Bioorg Med Chem ; 19(8): 2589-95, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21459001

ABSTRACT

The pharmacological suppression of the DNA damage response and DNA repair can increase the therapeutic indices of conventional chemotherapeutics. Replication Protein A (RPA), the major single-stranded DNA binding protein in eukaryotes, is required for DNA replication, DNA repair, DNA recombination, and DNA damage response signaling. Through the use of high-throughput screening of 1500 compounds, we have identified a small molecule inhibitor, 15-carboxy-13-isopropylatis-13-ene-17,18-dioic acid (NSC15520), that inhibited both the binding of Rad9-GST and p53-GST fusion proteins to the RPA N-terminal DNA binding domain (DBD), interactions that are essential for robust DNA damage signaling. NSC15520 competitively inhibited the binding of p53-GST peptide with an IC(50) of 10 µM. NSC15520 also inhibited helix destabilization of a duplex DNA (dsDNA) oligonucleotide, an activity dependent on the N-terminal domain of RPA70. NSC15520 did not inhibit RPA from binding single-stranded oligonucleotides, suggesting that the action of this inhibitor is specific for the N-terminal DBD of RPA, and does not bind to DBDs essential for single-strand DNA binding. Computer modeling implicates direct competition between NSC15520 and Rad9 for the same binding surface on RPA. Inhibitors of protein-protein interactions within the N-terminus of RPA are predicted to act synergistically with DNA damaging agents and inhibitors of DNA repair. Novel compounds such as NSC15520 have the potential to serve as chemosensitizing agents.


Subject(s)
Protein Interaction Domains and Motifs/drug effects , Replication Protein A/antagonists & inhibitors , Binding Sites , Computer Simulation , DNA Repair , DNA, Single-Stranded , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Humans , Oligonucleotides
18.
Nucleic Acids Res ; 38(3): 846-58, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19942684

ABSTRACT

Replication Protein A (RPA) is a single-stranded DNA-binding protein essential for DNA replication, repair, recombination and cell-cycle regulation. A human homolog of the RPA2 subunit, called RPA4, was previously identified and shown to be expressed in colon mucosal and placental cells; however, the function of RPA4 was not determined. To examine the function of RPA4 in human cells, we carried out knockdown and replacement studies to determine whether RPA4 can substitute for RPA2 in the cell. Unlike RPA2, exogenous RPA4 expression did not support chromosomal DNA replication and lead to cell-cycle arrest in G2/M. In addition, RPA4 localized to sites of DNA repair and reduced gamma-H2AX caused by RPA2 depletion. These studies suggest that RPA4 cannot support cell proliferation but can support processes that maintain the genomic integrity of the cell.


Subject(s)
Cell Cycle , DNA Replication , DNA-Binding Proteins/metabolism , Amino Acid Sequence , Apoptosis , DNA Repair , DNA-Binding Proteins/chemistry , Genomics , HeLa Cells , Humans , Molecular Sequence Data , Phenotype , Protein Subunits/metabolism , Replication Protein A/antagonists & inhibitors , Replication Protein A/metabolism , Sequence Homology, Amino Acid
19.
Oncol Rep ; 19(2): 535-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202804

ABSTRACT

The polyamines are aliphatic cations which are present in millimolar concentrations in all mammalian cells, and are required for optimal growth of almost all cell types. In this study, the roles of polyamines in DNA replication in vitro and the mechanism by which polyamines affected DNA replication were examined using simian virus 40 DNA replication system in vitro. We found that polyamines inhibited DNA replication, but it is not clear at which stage this occurs. Spermidine inhibited the DNA cleavage by topoisomerase I at 8.0 mM, but stimulated its activity at 1.0 mM. Spermine also inhibited its activity at 4.0 mM, but stimulated at 1.0 mM. The ssDNA binding activity of replication protein A was slightly affected by polyamines. Polyamines, especially spermine, also significantly reduced polymerase alpha-primase activity at 133 microM. Taken together, we suggest that the major inhibition of SV40 DNA replication may be due to the inhibition of pol alpha-primase activity, and possible roles for polyamines in the initiation process are discussed.


Subject(s)
DNA Polymerase I/antagonists & inhibitors , DNA Primase/antagonists & inhibitors , DNA Replication , Spermidine/physiology , Spermine/physiology , DNA Cleavage/drug effects , DNA Replication/drug effects , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/metabolism , Humans , Polyamines/pharmacology , Replication Protein A/antagonists & inhibitors , Simian virus 40/drug effects , Simian virus 40/genetics , Spermidine/pharmacology , Spermine/pharmacology , Topoisomerase I Inhibitors
20.
Nucleic Acids Res ; 35(21): 7267-78, 2007.
Article in English | MEDLINE | ID: mdl-17959650

ABSTRACT

The activation of a telomere maintenance mechanism is required for cancer development in humans. While most tumors achieve this by expressing the enzyme telomerase, a fraction (5-15%) employs a recombination-based mechanism termed alternative lengthening of telomeres (ALT). Here we show that loss of the single-stranded DNA-binding protein replication protein A (RPA) in human ALT cells, but not in telomerase-positive cells, causes increased exposure of single-stranded G-rich telomeric DNA, cell cycle arrest in G2/M phase, accumulation of single-stranded telomeric DNA within ALT-associated PML bodies (APBs), and formation of telomeric aggregates at the ends of metaphase chromosomes. This study demonstrates differences between ALT cells and telomerase-positive cells in the requirement for RPA in telomere processing and implicates the ALT mechanism in tumor cells as a possible therapeutic target.


Subject(s)
DNA, Single-Stranded/metabolism , Neoplasms/genetics , Replication Protein A/physiology , Telomere/metabolism , Cell Cycle , Cell Line, Transformed , Cell Line, Tumor , Humans , RNA Interference , Replication Protein A/antagonists & inhibitors , Telomere/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...