Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(5): 1214-1215, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33822743

ABSTRACT

Defects in genes crucial for the process of DNA repair by homology-directed DNA repair (HDR), such as BRCA1 and BRCA2, are well-known contributors to cancer pathogenesis as well as an Achilles' heel that can be exploited therapeutically. BRCA1/2-deficient cells are exquisitely sensitive to agents that stall replication forks, such as PARP inhibitors and platinating drugs, presumably due to the inability to repair double-stranded breaks that form as a consequence of replication fork collapse. BRCA1/2 also promote tolerance to DNA replication stress by protecting replication forks from nucleolytic degradation. Both biological endpoints involve the deposition of RAD51 onto single-stranded DNA (ssDNA) for homology searching and strand exchange during HDR repair, as well as protection of newly synthesized DNA from nucleolytic degradation (i.e., replication fork protection). In this issue of Cancer Research, Panzarino and colleagues performed multiple separation-of-function studies and identify the lesion most associated with intolerance to replication stress in BRCA1/2-deficient cells is persistent ssDNA gaps in newly synthesized DNA, resulting from a failure to restrain DNA replication. Mechanisms that suppress gap formation are closely associated with chemoresistance, and the authors' findings challenge the paradigm that lack of HR repair or fork protection underlie the phenotype known as BRCAness.See related article by Panzarino et al., p. 1388.


Subject(s)
Pharmaceutical Preparations , Rad51 Recombinase , DNA Repair/genetics , DNA Replication , Genomic Instability , Humans , Rad51 Recombinase/metabolism
2.
Mol Cancer Ther ; 20(2): 263-273, 2021 02.
Article in English | MEDLINE | ID: mdl-33268569

ABSTRACT

PARP inhibitor monotherapy (olaparib) was recently FDA approved for the treatment of BRCA1/2-mutant, homologous recombination (HR) repair-deficient pancreatic cancer. Most pancreatic cancers, however, are HR proficient and thus resistant to PARP inhibitor monotherapy. We tested the hypothesis that combined therapy with radiation and ataxia telangiectasia and Rad3-related (ATR) inhibitor (AZD6738) would extend the therapeutic indication of olaparib to HR-proficient pancreatic cancers. We show that olaparib combined with AZD6738 significantly reduced radiation survival relative to either agent alone, regardless of HR status. Whereas catalytic inhibition of PARP with low concentrations of olaparib radiosensitized HR-deficient models, maximal sensitization in HR-proficient models required concentrations of olaparib that induce formation of PARP1-DNA complexes. Furthermore, CRISPR-Cas9-mediated PARP1 deletion failed to recapitulate the effects of olaparib on radiosensitivity and negated the combinatorial efficacy of olaparib and AZD6738 on radiosensitization, suggesting that PARP1-DNA complexes, rather than PARP catalytic inhibition, were responsible for radiosensitization. Mechanistically, therapeutic concentrations of olaparib in combination with radiation and AZD6738 increased DNA double-strand breaks. DNA fiber combing revealed that high concentrations of olaparib did not stall replication forks but instead accelerated replication fork progression in association with an ATR-mediated replication stress response that was antagonized by AZD6738. Finally, in HR-proficient tumor xenografts, the combination of olaparib, radiation, and AZD6738 significantly delayed tumor growth compared with all other treatments. These findings suggest that PARP1-DNA complexes are required for the therapeutic activity of olaparib combined with radiation and ATR inhibitor in HR-proficient pancreatic cancer and support the clinical development of this combination for tumors intrinsically resistant to PARP inhibitors.


Subject(s)
Combined Modality Therapy/methods , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Animals , Humans , Male , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pancreatic Neoplasms
3.
Cancer Res ; 78(24): 6713-6716, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30498082

ABSTRACT

Glioblastoma (GBM) is a highly aggressive form of cancer that is resistant to standard therapy with concurrent radiation and temozolomide, two agents that work by inducing DNA damage. An underlying cause of this resistance may be a subpopulation of cancer stem-like cells that display a heightened DNA damage response (DDR). Although this DDR represents an attractive therapeutic target for overcoming the resistance of GBMs to radiotherapy, until now, the cause of this DDR upregulation has not been understood. In a previous issue of Cancer Research, Carruthers and colleagues investigated DNA replication stress as an underlying mechanism responsible for upregulation of the DDR and hence the radiation resistance of glioma stem-like cells. Furthermore, the authors explore the efficacy of combined ataxia telangiectasia and Rad3-related kinase and PARP inhibitors as a strategy to leverage these mechanisms and overcome radiation resistance.See related article by Carruthers and colleagues, Cancer Res; 78(17); 5060-71.


Subject(s)
Glioblastoma , Glioma , Cell Line, Tumor , DNA Damage , Humans , Neoplastic Stem Cells
4.
DNA Repair (Amst) ; 46: 37-46, 2016 10.
Article in English | MEDLINE | ID: mdl-27481099

ABSTRACT

Rev3, the catalytic subunit of yeast DNA polymerase ζ, is required for UV resistance and UV-induced mutagenesis, while its mammalian ortholog, REV3L, plays further vital roles in cell proliferation and embryonic development. To assess the contribution of REV3L catalytic activity to its in vivo function, we generated mutant mouse strains in which one or two Ala residues were substituted to the Asp of the invariant catalytic YGDTDS motif. The simultaneous mutation of both Asp (ATA) phenocopies the Rev3l knockout, which proves that the catalytic activity is mandatory for the vital functions of Rev3L, as reported recently. Surprisingly, although the mutation of the first Asp severely impairs the enzymatic activity of other B-family DNA polymerases, the corresponding mutation of Rev3 (ATD) is hypomorphic in yeast and mouse, as it does not affect viability and proliferation and moderately impacts UVC-induced cell death and mutagenesis. Interestingly, Rev3l hypomorphic mutant mice display a distinct, albeit modest, alteration of the immunoglobulin gene mutation spectrum at G-C base pairs, further documenting its role in this process.


Subject(s)
Aspartic Acid/metabolism , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Mutation , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Catalytic Domain , Cell Line , Cell Survival/radiation effects , Conserved Sequence , DNA-Binding Proteins/deficiency , DNA-Directed DNA Polymerase/deficiency , DNA-Directed DNA Polymerase/metabolism , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , HEK293 Cells , Humans , Immunoglobulins/genetics , Mice , Mice, Transgenic , Phenotype , Ultraviolet Rays
5.
Nat Commun ; 5: 5437, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25421715

ABSTRACT

Cells cope with replication-blocking lesions via translesion DNA synthesis (TLS). TLS is carried out by low-fidelity DNA polymerases that replicate across lesions, thereby preventing genome instability at the cost of increased point mutations. Here we perform a two-stage siRNA-based functional screen for mammalian TLS genes and identify 17 validated TLS genes. One of the genes, NPM1, is frequently mutated in acute myeloid leukaemia (AML). We show that NPM1 (nucleophosmin) regulates TLS via interaction with the catalytic core of DNA polymerase-η (polη), and that NPM1 deficiency causes a TLS defect due to proteasomal degradation of polη. Moreover, the prevalent NPM1c+ mutation that causes NPM1 mislocalization in ~30% of AML patients results in excessive degradation of polη. These results establish the role of NPM1 as a key TLS regulator, and suggest a mechanism for the better prognosis of AML patients carrying mutations in NPM1.


Subject(s)
DNA Damage , DNA Replication , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Cell Line , DNA Damage/radiation effects , DNA Repair , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , Nucleophosmin , Protein Binding , Ultraviolet Rays
6.
J Biol Chem ; 289(10): 7109-7120, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24474685

ABSTRACT

Small molecule inhibitors of proliferating cell nuclear antigen (PCNA)/PCNA interacting protein box (PIP-Box) interactions, including T2 amino alcohol (T2AA), inhibit translesion DNA synthesis. The crystal structure of PCNA in complex with T2AA revealed that T2AA bound to the surface adjacent to the subunit interface of the homotrimer of PCNA in addition to the PIP-box binding cavity. Because this site is close to Lys-164, which is monoubiquitinated by RAD18, we postulated that T2AA would affect monoubiquitinated PCNA interactions. Binding of monoubiquitinated PCNA and a purified pol η fragment containing the UBZ and PIP-box was inhibited by T2AA in vitro. T2AA decreased PCNA/pol η and PCNA/REV1 chromatin colocalization but did not inhibit PCNA monoubiquitination, suggesting that T2AA hinders interactions of pol η and REV1 with monoubiquitinated PCNA. Interstrand DNA cross-links (ICLs) are repaired by mechanisms using translesion DNA synthesis that is regulated by monoubiquitinated PCNA. T2AA significantly delayed reactivation of a reporter plasmid containing an ICL. Neutral comet analysis of cells receiving T2AA in addition to cisplatin revealed that T2AA significantly enhanced formation of DNA double strand breaks (DSBs) by cisplatin. T2AA promoted colocalized foci formation of phospho-ATM and 53BP1 and up-regulated phospho-BRCA1 in cisplatin-treated cells, suggesting that T2AA increases DSBs. When cells were treated by cisplatin and T2AA, their clonogenic survival was significantly less than that of those treated by cisplatin only. These findings show that the inhibitors of monoubiquitinated PCNA chemosensitize cells by inhibiting repair of ICLs and DSBs.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/metabolism , Phenyl Ethers/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Propanolamines/pharmacology , Small Molecule Libraries/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Crystallography, X-Ray , HeLa Cells , Humans , Neoplasms/genetics , Phenyl Ethers/chemistry , Proliferating Cell Nuclear Antigen/chemistry , Propanolamines/chemistry
7.
Cancer Res ; 74(6): 1778-88, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24469230

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by therapeutic resistance for which the basis is poorly understood. Here, we report that the DNA and p53-binding protein ATDC/TRIM29, which is highly expressed in PDAC, plays a critical role in DNA damage signaling and radioresistance in pancreatic cancer cells. Ataxia-telangiectasia group D-associated gene (ATDC) mediated resistance to ionizing radiation in vitro and in vivo in mouse xenograft assays. ATDC was phosphorylated directly by MAPKAP kinase 2 (MK2) at Ser550 in an ATM-dependent manner. Phosphorylation at Ser-550 by MK2 was required for the radioprotective function of ATDC. Our results identify a DNA repair pathway leading from MK2 and ATM to ATDC, suggesting its candidacy as a therapeutic target to radiosensitize PDAC and improve the efficacy of DNA-damaging treatment.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Survival/radiation effects , DNA-Binding Proteins/genetics , Dishevelled Proteins , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/radiotherapy , Phosphoproteins/metabolism , Phosphorylation , Radiation Tolerance , Transcription Factors/genetics , Xenograft Model Antitumor Assays
8.
Cancer Res ; 74(6): 1651-60, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24473064

ABSTRACT

Impairment of double-stranded DNA break (DSB) repair is essential to many cancers. However, although mutations in DSB repair proteins are common in hereditary cancers, mechanisms of impaired DSB repair in sporadic cancers remain incompletely understood. Here, we describe the first role for a long noncoding RNA (lncRNA) in DSB repair in prostate cancer. We identify PCAT-1, a prostate cancer outlier lncRNA, which regulates cell response to genotoxic stress. PCAT-1 expression produces a functional deficiency in homologous recombination through its repression of the BRCA2 tumor suppressor, which, in turn, imparts a high sensitivity to small-molecule inhibitors of PARP1. These effects reflected a posttranscriptional repression of the BRCA2 3'UTR by PCAT-1. Our observations thus offer a novel mechanism of "BRCAness" in sporadic cancers.


Subject(s)
BRCA2 Protein/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Recombinational DNA Repair , 3' Untranslated Regions , Animals , Antineoplastic Agents/pharmacology , BRCA2 Protein/metabolism , Cell Death/drug effects , Cell Line, Tumor , DNA Damage , Humans , Male , Mice , Mice, SCID , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Long Noncoding/metabolism , Xenograft Model Antitumor Assays
9.
DNA Repair (Amst) ; 12(12): 1114-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24231389

ABSTRACT

The nucleoside analog ganciclovir (GCV) elicits cytotoxicity in tumor cells via a novel mechanism in which drug incorporation into DNA produces minimal disruption of replication, but numerous DNA double strand breaks occur during the second S-phase after drug exposure. We propose that homologous recombination (HR), a major repair pathway for DNA double strand breaks, can prevent GCV-induced DNA damage, and that inhibition of HR will enhance cytotoxicity with GCV. Survival after GCV treatment in cells expressing a herpes simplex virus thymidine kinase was strongly dependent on HR (>14-fold decrease in IC50 in HR-deficient vs. HR-proficient CHO cells). In a homologous recombination reporter assay, the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA; vorinostat), decreased HR repair events up to 85%. SAHA plus GCV produced synergistic cytotoxicity in U251tk human glioblastoma cells. Elucidation of the synergistic mechanism demonstrated that SAHA produced a concentration-dependent decrease in the HR proteins Rad51 and CtIP. GCV alone produced numerous Rad51 foci, demonstrating activation of HR. However, the addition of SAHA blocked GCV-induced Rad51 foci formation completely and increased γH2AX, a marker of DNA double strand breaks. SAHA plus GCV also produced synergistic cytotoxicity in HR-proficient CHO cells, but the combination was antagonistic or additive in HR-deficient CHO cells. Collectively, these data demonstrate that HR promotes survival with GCV and compromise of HR by SAHA results in synergistic cytotoxicity, revealing a new mechanism for enhancing anticancer activity with GCV.


Subject(s)
Antineoplastic Agents/pharmacology , Ganciclovir/pharmacology , Homologous Recombination/drug effects , Hydroxamic Acids/pharmacology , Animals , Apoptosis/drug effects , CHO Cells , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cricetulus , Endodeoxyribonucleases , HeLa Cells , Humans , Nuclear Proteins/metabolism , Rad51 Recombinase/metabolism , Vorinostat
10.
Cell Cycle ; 12(24): 3749-58, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24107634

ABSTRACT

The rapid ubiquitination of chromatin surrounding DNA double-stranded breaks (DSB) drives the formation of large structures called ionizing radiation-induced foci (IRIF), comprising many DNA damage response (DDR) proteins. This process is regulated by RNF8 and RNF168 ubiquitin ligases and is thought to be necessary for DNA repair and activation of signaling pathways involved in regulating cell cycle checkpoints. Here we demonstrate that it is possible to interfere with ubiquitin-dependent recruitment of DDR factors by expressing proteins containing ubiquitin binding domains (UBDs) that bind to lysine 63-linked polyubiquitin chains. Expression of the E3 ubiquitin ligase RAD18 prevented chromatin spreading of 53BP1 at DSBs, and this phenomenon was dependent upon the integrity of the RAD18 UBD. An isolated RAD18 UBD interfered with 53BP1 chromatin spreading, as well as other important DDR mediators, including RAP80 and the BRCA1 tumor suppressor protein, consistent with the model that the RAD18 UBD is blocking access of proteins to ubiquitinated chromatin. Using the RAD18 UBD as a tool to impede localization of 53BP1 and BRCA1 to repair foci, we found that DDR signaling, DNA DSB repair, and radiosensitivity were unaffected. We did find that activated ATM (S1981P) and phosphorylated SMC1 (a specific target of ATM) were not detectable in DNA repair foci, in addition to upregulated homologous recombination repair, revealing 2 DDR responses that are dependent upon chromatin spreading of certain DDR factors at DSBs. These data demonstrate that select UBDs containing targeting motifs may be useful probes in determining the biological significance of protein-ubiquitin interactions.


Subject(s)
DNA Repair , Ubiquitin/metabolism , BRCA1 Protein/metabolism , Carrier Proteins/metabolism , Cell Line , Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Histone Chaperones , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Radiation Tolerance , Recombinational DNA Repair , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin/genetics , Ubiquitin-Protein Ligases , Ubiquitination
11.
Proc Natl Acad Sci U S A ; 110(33): 13558-63, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901102

ABSTRACT

Breast cancer gene 1 (BRCA1) deficient cells not only are hypersensitive to double-strand breaks but also are hypersensitive to UV irradiation and other agents that cause replication blockade; however, the molecular mechanisms behind these latter sensitivities are largely unknown. Here, we report that BRCA1 promotes cell survival by directly regulating the DNA damage tolerance pathway in response to agents that create cross-links in DNA. We show that BRCA1 not only promotes efficient mono- and polyubiquitination of proliferating cell nuclear antigen (PCNA) by regulating the recruitment of replication protein A, Rad18, and helicase-like transcription factor to chromatin but also directly recruits translesion polymerases, such as Polymerase eta and Rev1, to the lesions through protein-protein interactions. Our data suggest that BRCA1 plays a critical role in promoting translesion DNA synthesis as well as DNA template switching.


Subject(s)
BRCA1 Protein/metabolism , Cell Survival/physiology , DNA Damage/physiology , Proliferating Cell Nuclear Antigen/metabolism , BRCA1 Protein/physiology , Chromatin/metabolism , Cross-Linking Reagents/toxicity , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Plasmids/genetics , RNA, Small Interfering/genetics , Replication Protein A/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases , Ubiquitination
12.
Mutat Res ; 743-744: 97-110, 2013.
Article in English | MEDLINE | ID: mdl-23195997

ABSTRACT

Cancer cells display numerous abnormal characteristics which are initiated and maintained by elevated mutation rates and genome instability. Chromosomal DNA is continuously surveyed for the presence of damage or blocked replication forks by the DNA Damage Response (DDR) network. The DDR is complex and includes activation of cell cycle checkpoints, DNA repair, gene transcription, and induction of apoptosis. Duplicating a damaged genome is associated with elevated risks to fork collapse and genome instability. Therefore, the DNA damage tolerance (DDT) pathway is also employed to enhance survival and involves the recruitment of translesion DNA synthesis (TLS) polymerases to sites of replication fork blockade or single stranded DNA gaps left after the completion of replication in order to restore DNA to its double stranded form before mitosis. TLS polymerases are specialized for inserting nucleotides opposite DNA adducts, abasic sites, or DNA crosslinks. By definition, the DDT pathway is not involved in the actual repair of damaged DNA, but provides a mechanism to tolerate DNA lesions during replication thereby increasing survival and lessening the chance for genome instability. However this may be associated with increased mutagenesis. In this review, we will describe the specialized functions of Y family polymerases (Rev1, Polη, Polι and Polκ) and DNA polymerase ζ in lesion bypass, mutagenesis, and prevention of genome instability, the latter due to newly appreciated roles in DNA repair. The recently described role of the Fanconi anemia pathway in regulating Rev1 and Polζ-dependent TLS is also discussed in terms of their involvement in TLS, interstrand crosslink repair, and homologous recombination.


Subject(s)
DNA Damage , DNA Repair , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Genomic Instability , Animals , DNA Replication/genetics , Humans
13.
Environ Mol Mutagen ; 53(9): 725-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065650

ABSTRACT

DNA interstrand crosslinks (ICLs) are covalent linkages between two strands of DNA, and their presence interferes with essential metabolic processes such as transcription and replication. These lesions are extremely toxic, and their repair is essential for genome stability and cell survival. In this review, we will discuss how the removal of ICLs requires interplay between multiple genome maintenance pathways and can occur in the absence of replication (replication-independent ICL repair) or during S phase (replication-coupled ICL repair), the latter being the predominant pathway used in mammalian cells. It is now well recognized that translesion DNA synthesis (TLS), especially through the activities of REV1 and DNA polymerase zeta (Polζ), is necessary for both ICL repair pathways operating throughout the cell cycle. Recent studies suggest that the convergence of two replication forks upon an ICL initiates a cascade of events including unhooking of the lesion through the actions of structure-specific endonucleases, thereby creating a DNA double-stranded break (DSB). TLS across the unhooked lesion is necessary for restoring the sister chromatid before homologous recombination repair. Biochemical and genetic studies implicate REV1 and Polζ as being essential for performing lesion bypass across the unhooked crosslink, and this step appears to be important for subsequent events to repair the intermediate DSB. The potential role of Fanconi anemia pathway in the regulation of REV1 and Polζ-dependent TLS and the involvement of additional polymerases, including DNA polymerases kappa, nu, and theta, in the repair of ICLs is also discussed in this review.


Subject(s)
DNA Repair , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , DNA Damage , DNA-Directed DNA Polymerase/metabolism , Mutagenesis , Recombination, Genetic
14.
Mol Pharmacol ; 81(6): 778-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22387291

ABSTRACT

Oxaliplatin, satraplatin, and picoplatin are cisplatin analogs that interact with DNA forming intrastrand and interstrand DNA cross-links (ICLs). Replicative bypass of cisplatin DNA adducts requires the cooperative actions of at least three translesion DNA synthesis (TLS) polymerases: Polη, REV1, and Polζ. Because oxaliplatin, satraplatin, and picoplatin contain bulkier chemical groups attached to the platinum core compared with cisplatin, we hypothesized that these chemical additions may impede replicative bypass by TLS polymerases and reduce tolerance to platinum-containing adducts. We examined multiple responses of cancer cells to oxaliplatin, satraplatin, or picoplatin treatment under conditions where expression of a TLS polymerase was limited. Our studies revealed that, although Polη contributes to the tolerance of cisplatin adducts, it plays a lesser role in promoting replication through oxaliplatin, satraplatin, and picoplatin adducts. REV1 and Polζ were necessary for tolerance to all four platinum analogs and prevention of hyperactivation of the DNA damage response after treatment. In addition, REV1 and Polζ were important for the resolution of DNA double-stranded breaks created during replication-associated repair of platinum-containing ICLs. Consistent with ICLs being the predominant cytotoxic lesion, depletion of REV1 or Polζ rendered two different model cell systems extremely sensitive to all four drugs, whereas Polη depletion had little effect. Together, our data suggest that REV1 and Polζ are critical for promoting resistance to all four clinically relevant platinum-based drugs by promoting both translesion DNA synthesis and DNA repair.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Organoplatinum Compounds/pharmacology , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Adducts , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , RNA, Small Interfering
15.
Nucleic Acids Res ; 40(2): 682-91, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21926160

ABSTRACT

REV1 and DNA Polymerase ζ (REV3 and REV7) play important roles in translesion DNA synthesis (TLS) in which DNA replication bypasses blocking lesions. REV1 and Polζ have also been implicated in promoting repair of DNA double-stranded breaks (DSBs). However, the mechanism by which these two TLS polymerases increase tolerance to DSBs is poorly understood. Here we demonstrate that full-length human REV1, REV3 and REV7 interact in vivo (as determined by co-immunoprecipitation studies) and together, promote homologous recombination repair. Cells lacking REV3 were hypersensitive to agents that cause DSBs including the PARP inhibitor, olaparib. REV1, REV3 or REV7-depleted cells displayed increased chromosomal aberrations, residual DSBs and sites of HR repair following exposure to ionizing radiation. Notably, cells depleted of DNA polymerase η (Polη) or the E3 ubiquitin ligase RAD18 were proficient in DSB repair following exposure to IR indicating that Polη-dependent lesion bypass or RAD18-dependent monoubiquitination of PCNA are not necessary to promote REV1 and Polζ-dependent DNA repair. Thus, the REV1/Polζ complex maintains genomic stability by directly participating in DSB repair in addition to the canonical TLS pathway.


Subject(s)
DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Proteins/metabolism , Recombinational DNA Repair , Cells, Cultured , Chromosomal Instability , DNA Breaks, Double-Stranded , DNA-Binding Proteins/physiology , DNA-Directed DNA Polymerase/physiology , Humans , Mad2 Proteins , Nuclear Proteins/physiology , Nucleotidyltransferases/physiology , Proteins/physiology , Radiation Tolerance , Radiation, Ionizing
16.
Cancer Res ; 70(12): 4972-81, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501833

ABSTRACT

The median survival for patients with locally advanced pancreatic cancer treated with gemcitabine and radiation is approximately 1 year. To develop improved treatment, we have combined a Chk1/2-targeted agent, AZD7762, currently in phase I clinical trials, with gemcitabine and ionizing radiation in preclinical pancreatic tumor models. We found that in vitro AZD7762 alone or in combination with gemcitabine significantly sensitized MiaPaCa-2 cells to radiation. AZD7762 inhibited Chk1 autophosphorylation (S296 Chk1), stabilized Cdc25A, and increased ATR/ATM-mediated Chk1 phosphorylation (S345 Chk1). Radiosensitization by AZD7762 was associated with abrogation of the G(2) checkpoint as well as with inhibition of Rad51 focus formation, inhibition of homologous recombination repair, and persistent gamma-H2AX expression. AZD7762 was also a radiation sensitizer in multiple tumor xenograft models. In both MiaPaCa-2- and patient-derived xenografts, AZD7762 significantly prolonged the median time required for tumor volume doubling in response to gemcitabine and radiation. Together, our findings suggest that G(2) checkpoint abrogation and homologous recombination repair inhibition both contribute to sensitization by Chk1 inhibition. Furthermore, they support the clinical use of AZD7762 in combination with gemcitabine and radiation for patients with locally advanced pancreatic cancer.


Subject(s)
DNA Repair/drug effects , G2 Phase/drug effects , Pancreatic Neoplasms/radiotherapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Thiophenes/pharmacology , Urea/analogs & derivatives , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Blotting, Western , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Therapy, Combination , Flow Cytometry , Fluorescent Antibody Technique , G2 Phase/radiation effects , Gamma Rays , Humans , Immunoblotting , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Kinases/chemistry , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rad51 Recombinase/metabolism , Recombination, Genetic/drug effects , Recombination, Genetic/radiation effects , Reverse Transcriptase Polymerase Chain Reaction , Urea/pharmacology , Xenograft Model Antitumor Assays , Gemcitabine
17.
Mol Cell Biol ; 30(5): 1217-30, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028736

ABSTRACT

Translesion DNA synthesis (TLS) is a process whereby specialized DNA polymerases are recruited to bypass DNA lesions that would otherwise stall high-fidelity polymerases. We provide evidence that TLS across cisplatin intrastrand cross-links is performed by multiple translesion DNA polymerases. First, we determined that PCNA monoubiquitination by RAD18 is necessary for efficient bypass of cisplatin adducts by the TLS polymerases eta (Poleta), REV1, and zeta (Polzeta) based on the observations that depletion of these proteins individually leads to decreased cell survival, cell cycle arrest in S phase, and activation of the DNA damage response. Second, we showed that in addition to PCNA monoubiquitination by RAD18, the Fanconi anemia core complex is also important for recruitment of REV1 to stalled replication forks in cisplatin treated cells. Third, we present evidence that REV1 and Polzeta are uniquely associated with protection against cisplatin and mitomycin C-induced chromosomal aberrations, and both are necessary for the timely resolution of DNA double-strand breaks associated with repair of DNA interstrand cross-links. Together, our findings indicate that REV1 and Polzeta facilitate repair of interstrand cross-links independently of PCNA monoubiquitination and Poleta, whereas RAD18 plus Poleta, REV1, and Polzeta are all necessary for replicative bypass of cisplatin intrastrand DNA cross-links.


Subject(s)
DNA Repair/physiology , DNA-Directed DNA Polymerase/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Base Sequence , Cell Cycle , Cell Line , Chromosome Aberrations , Cisplatin/toxicity , Cross-Linking Reagents/toxicity , DNA/chemistry , DNA/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/genetics , HeLa Cells , Humans , Mad2 Proteins , Mitomycin/toxicity , Models, Biological , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nucleic Acid Synthesis Inhibitors , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proteins/antagonists & inhibitors , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , Ubiquitin-Protein Ligases
18.
Mol Cancer Ther ; 8(1): 45-54, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19139112

ABSTRACT

The protein kinase checkpoint kinase 1 (Chk1) has been implicated as a key regulator of cell cycle progression and DNA repair, and inhibitors of Chk1 (e.g., UCN-01 and EXEL-9844) potentiate the cytotoxic actions of chemotherapeutic drugs in tumor cells. We have examined the ability of PD-321852, a small-molecule Chk1 inhibitor, to potentiate gemcitabine-induced clonogenic death in a panel of pancreatic cancer cell lines and evaluated the relationship between endpoints associated with Chk1 inhibition and chemosensitization. Gemcitabine chemosensitization by minimally toxic concentrations of PD-321852 ranged from minimal (<3-fold change in survival) in Panc1 cells to >30-fold in MiaPaCa2 cells. PD-321852 inhibited Chk1 in all cell lines as evidenced by stabilization of Cdc25A; in combination with gemcitabine, a synergistic loss of Chk1 protein was observed in the more sensitized cell lines. Gemcitabine chemosensitization, however, did not correlate with abrogation of the S-M or G2-M checkpoint; PD-321852 did not induce premature mitotic entry in gemcitabine-treated BxPC3 or M-Panc96 cells, which were sensitized to gemcitabine 6.2- and 4.6-fold, respectively. In the more sensitized cells lines, PD-321852 not only inhibited gemcitabine-induced Rad51 focus formation and the recovery from gemcitabine-induced replication stress, as evidenced by persistence of gamma-H2AX, but also depleted these cells of Rad51 protein. Our data suggest the inhibition of this Chk1-mediated Rad51 response to gemcitabine-induced replication stress is an important factor in determining gemcitabine chemosensitization by Chk1 inhibition in pancreatic cancer cells.


Subject(s)
Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Biocatalysis , Carbazoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Checkpoint Kinase 1 , DNA Damage , Deoxycytidine/pharmacology , Humans , Pancreatic Neoplasms/genetics , Phosphorylation/drug effects , Gemcitabine
19.
Mol Pharmacol ; 75(3): 599-607, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19064630

ABSTRACT

Psoralen plus UVA light (PUVA) is commonly used to treat psoriasis, a common skin disorder associated with rapid proliferation of cells. PUVA exerts its antiproliferative activity through formation of DNA monoadducts and interstrand cross-links (ICLs). However, this treatment may lead to skin malignancies as a direct result of inducing carcinogenic DNA damage. Inactivation of the p53 tumor suppressor gene is an important event in the development of skin cancer. p53 is rapidly phosphorylated and stabilized in response to DNA damage, and the induction of apoptosis by p53 is an important mechanism by which p53 exerts its tumor-suppressive activity. To better understand the mechanism by which PUVA treatment induces p53, we exposed human skin fibroblasts with PUVA under conditions that differentially produce monoadducts and ICLs and found that psoralen-induced ICLs induced phosphorylation of the Ser-15 site of p53 and apoptosis much more effectively than psoralen-induced monoadducts. The induction of p53 phosphorylation by psoralen ICLs did not require factors believed to be involved in the repair of psoralen ICLs [xeroderma pigmentosum (XP)-A, XP-C, XP-F, Cockayne's syndrome-B, Fanconi anemia] but did require the ataxia-telangiectasia and Rad3-related but not the ataxia-telangiectasia mutated kinase. Psoralen-induced ICLs blocked transcription and replication more efficiently than monoadducts, and induction of p53 and apoptosis correlated with doses causing interference with transcription rather than DNA replication. Our finding that cells underwent apoptosis preferentially during S-phase suggests that the combined blockade of transcription and DNA replication by psoralen ICLs during S-phase elicits a strong apoptotic response.


Subject(s)
Ataxia Telangiectasia , Cell Cycle Proteins/physiology , Cross-Linking Reagents/toxicity , DNA Damage/drug effects , Ficusin/toxicity , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/biosynthesis , Ataxia Telangiectasia/chemically induced , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Line, Transformed , DNA Damage/radiation effects , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays/adverse effects
20.
Cancer Res ; 66(24): 11554-9, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178844

ABSTRACT

Ataxia telangiectasia mutated (ATM) kinase plays a crucial role in the cellular response to DNA damage and in radiation resistance. Although much effort has focused on the relationship between ATM and other nuclear signal transducers, little is known about interactions between ATM and mitogenic signaling pathways. In this study, we show a novel relationship between ATM kinase and extracellular signal-regulated kinase 1/2 (ERK1/2), a key mitogenic stimulator. Activation of ATM by radiation down-regulates phospho-ERK1/2 and its downstream signaling via increased expression of mitogen-activated protein kinase phosphatase MKP-1 in both cell culture and tumor models. This dephosphorylation of ERK1/2 is independent of epidermal growth factor receptor (EGFR) activity and is associated with radioresistance. These findings show a new function for ATM in the control of mitogenic pathways affecting cell signaling and emphasize the key role of ATM in coordinating the cellular response to DNA damage.


Subject(s)
Antigens, Differentiation/genetics , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/radiation effects , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/radiation effects , Membrane Glycoproteins/genetics , Neural Cell Adhesion Molecules/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/radiation effects , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/radiation effects , Receptors, Immunologic/genetics , Tumor Suppressor Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Survival , DNA Damage , DNA Replication , Dual Specificity Phosphatase 1 , Enzyme Activation , Humans , Mice , Mice, Nude , Polymerase Chain Reaction , Protein Phosphatase 1 , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...