151
|
Guainazzi A, Schärer OD. Using synthetic DNA interstrand crosslinks to elucidate repair pathways and identify new therapeutic targets for cancer chemotherapy. Cell Mol Life Sci 2010; 67:3683-97. [PMID: 20730555 PMCID: PMC3732395 DOI: 10.1007/s00018-010-0492-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 07/28/2010] [Indexed: 01/16/2023]
Abstract
Many cancer chemotherapeutic agents form DNA interstrand crosslinks (ICLs), extremely cytotoxic lesions that form covalent bonds between two opposing DNA strands, blocking DNA replication and transcription. However, cellular responses triggered by ICLs can cause resistance in tumor cells, limiting the efficacy of such treatment. Here we discuss recent advances in our understanding of the mechanisms of ICL repair that cause this resistance. The recent development of strategies for the synthesis of site-specific ICLs greatly contributed to these insights. Key features of repair are similar for all ICLs, but there is increasing evidence that the specifics of lesion recognition and synthesis past ICLs by DNA polymerases are dependent upon the structure of ICLs. These new insights provide a basis for the improvement of antitumor therapy by targeting DNA repair pathways that lead to resistance to treatment with crosslinking agents.
Collapse
Affiliation(s)
- Angelo Guainazzi
- Departments of Pharmacological Sciences, Chemistry 619, Stony Brook University, Stony Brook, NY 11794-3400 USA
| | - Orlando D. Schärer
- Departments of Pharmacological Sciences and Chemistry, Chemistry 619, Stony Brook University, Stony Brook, NY 11794-3400 USA
| |
Collapse
|
152
|
Hawtin RE, Stockett DE, Wong OK, Lundin C, Helleday T, Fox JA. Homologous recombination repair is essential for repair of vosaroxin-induced DNA double-strand breaks. Oncotarget 2010; 1:606-619. [PMID: 21317456 PMCID: PMC3248135 DOI: 10.18632/oncotarget.195] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 11/22/2010] [Indexed: 11/25/2022] Open
Abstract
Vosaroxin (formerly voreloxin) is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II, inducing site-selective double-strand breaks (DSB), G2 arrest and apoptosis. Objective responses and complete remissions were observed in phase 2 studies of vosaroxin in patients with solid and hematologic malignancies, and responses were seen in patients whose cancers were resistant to anthracyclines. The quinolone-based scaffold differentiates vosaroxin from the anthracyclines and anthracenediones, broadly used DNA intercalating topoisomerase II poisons. Here we report that vosaroxin induces a cell cycle specific pattern of DNA damage and repair that is distinct from the anthracycline, doxorubicin. Both drugs stall replication and preferentially induce DNA damage in replicating cells, with damage in G2 / M > S >> G1. However, detectable replication fork collapse, as evidenced by DNA fragmentation and long tract recombination during S phase, is induced only by doxorubicin. Furthermore, vosaroxin induces less overall DNA fragmentation. Homologous recombination repair (HRR) is critical for recovery from DNA damage induced by both agents, identifying the potential to clinically exploit synthetic lethality.
Collapse
Affiliation(s)
| | - David Elliot Stockett
- Sunesis Pharmaceuticals, Inc. 395 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Oi Kwan Wong
- Sunesis Pharmaceuticals, Inc. 395 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Cecilia Lundin
- Gray Institute for Radiation Oncology & Biology, University of Oxford. Old Road Campus Research Building, Roosevelt Drive. Oxford, OX3 7DQ, UK
| | - Thomas Helleday
- Gray Institute for Radiation Oncology & Biology, University of Oxford. Old Road Campus Research Building, Roosevelt Drive. Oxford, OX3 7DQ, UK
- Dept. of Genetics Microbiology and Toxicology, Stockholm University. Arrhenius Laboratory, Svante Arrhenius väg 16 E4. S-106 91 Stockholm, Sweden
| | - Judith Ann Fox
- Sunesis Pharmaceuticals, Inc. 395 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| |
Collapse
|
153
|
Formation of acetaldehyde-derived DNA adducts due to alcohol exposure. Chem Biol Interact 2010; 188:367-75. [PMID: 20813101 DOI: 10.1016/j.cbi.2010.08.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 08/18/2010] [Accepted: 08/25/2010] [Indexed: 12/12/2022]
Abstract
Epidemiological studies have identified chronic alcohol consumption as a significant risk factor for cancers of the upper aerodigestive tract, including the oral cavity, pharynx, larynx and esophagus, and for cancer of the liver. Ingested ethanol is mainly oxidized by the enzymes alcohol dehydrogenase (ADH), cytochrome P-450 2E1 (CYP2E1), and catalase to form acetaldehyde, which is subsequently oxidized by aldehyde dehydrogenase 2 (ALDH2) to produce acetate. Polymorphisms of the genes which encode enzymes for ethanol metabolism affect the ethanol/acetaldehyde oxidizing capacity. ADH1B*2 allele (ADH1B, one of the enzyme in ADH family) is commonly observed in Asian population, has much higher enzymatic activity than ADH1B*1 allele. Otherwise, approximately 40% of Japanese have single nucleotide polymorphisms (SNPs) of the ALDH2 gene. The ALDH2 *2 allele encodes a protein with an amino acid change from glutamate to lysine (derived from the ALDH2*1 allele) and devoid of enzymatic activity. Neither the homozygote (ALDH2*2/*2) nor heterozygote (ALDH2*1/*2) is able to metabolize acetaldehyde promptly. Acetaldehyde is a genotoxic compound that reacts with DNA to form primarily a Schiff base N(2)-ethylidene-2'-deoxyguanosine (N(2)-ethylidene-dG) adduct, which may be converted by reducing agents to N(2)-ethyl-2'-deoxyguanosine (N(2)-ethyl-dG) in vivo, and strongly blocked translesion DNA synthesis. Several studies have demonstrated a relationship between ALDH2 genotypes and the development of certain types of cancer. On the other hand, the drinking of alcohol induces the expression of CYP2E1, resulting in an increase in reactive oxygen species (ROS) and oxidative DNA damage. This review covers the combined effects of alcohol and ALDH2 polymorphisms on cancer risk. Studies show that ALDH2*1/*2 heterozygotes who habitually consume alcohol have higher rates of cancer than ALDH2*1/*1 homozygotes. Moreover, they support that chronic alcohol consumption contributes to formation of various DNA adducts. Although some DNA adducts formation is demonstrated to be an initiation step of carcinogenesis, it is still unclear that whether these alcohol-related DNA adducts are true factors or initiators of cancer. Future studies are needed to better characterize and to validate the roles of these DNA adducts in human study.
Collapse
|
154
|
Hartley JA, Hamaguchi A, Coffils M, Martin CRH, Suggitt M, Chen Z, Gregson SJ, Masterson LA, Tiberghien AC, Hartley JM, Pepper C, Lin TT, Fegan C, Thurston DE, Howard PW. SG2285, a novel C2-aryl-substituted pyrrolobenzodiazepine dimer prodrug that cross-links DNA and exerts highly potent antitumor activity. Cancer Res 2010; 70:6849-58. [PMID: 20660714 PMCID: PMC3533126 DOI: 10.1158/0008-5472.can-10-0790] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The pyrrolobenzodiazepines (PBD) are naturally occurring antitumor antibiotics, and a PBD dimer (SJG-136, SG2000) is in phase II trials. Many potent PBDs contain a C2-endo-exo unsaturated motif associated with the pyrrolo C-ring. The novel compound SG2202 is a PBD dimer containing this motif. SG2285 is a water-soluble prodrug of SG2202 in which two bisulfite groups inactivate the PBD N10-C11 imines. Once the bisulfites are eliminated, the imine moieties can bind covalently in the DNA minor groove, forming an interstrand cross-link. The mean in vitro cytotoxic potency of SG2285 against human tumor cell lines is GI(50) 20 pmol/L. SG2285 is highly efficient at producing DNA interstrand cross-links in cells, but they form more slowly than those produced by SG2202. Cellular sensitivity to SG2285 was primarily dependent on ERCC1 and homologous recombination repair. In primary B-cell chronic lymphocytic leukemia samples, the mean LD(50) was significantly lower than in normal age-matched B and T lymphocytes. Antitumor activity was shown in several human tumor xenograft models, including ovarian, non-small cell lung, prostate, pancreatic, and melanoma, with cures obtained in the latter model with a single dose. Further, in an advanced-stage colon model, SG2285 administered either as a single dose, or in two repeat dose schedules, was superior to irinotecan. Our findings define SG2285 as a highly active cytotoxic compound with antitumor properties desirable for further development.
Collapse
|
155
|
Gómez M, Serrano M, Pope CE, Jenkins J, Biancardi M, López M, Dumas C, Galiguis J, Dresser B. Derivation of cat embryonic stem-like cells from in vitro-produced blastocysts on homologous and heterologous feeder cells. Theriogenology 2010; 74:498-515. [DOI: 10.1016/j.theriogenology.2010.05.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 05/10/2010] [Accepted: 05/11/2010] [Indexed: 10/19/2022]
|
156
|
Kratz K, Schöpf B, Kaden S, Sendoel A, Eberhard R, Lademann C, Cannavó E, Sartori AA, Hengartner MO, Jiricny J. Deficiency of FANCD2-associated nuclease KIAA1018/FAN1 sensitizes cells to interstrand crosslinking agents. Cell 2010; 142:77-88. [PMID: 20603016 DOI: 10.1016/j.cell.2010.06.022] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 05/29/2010] [Accepted: 06/15/2010] [Indexed: 11/28/2022]
Abstract
Cytotoxicity of cisplatin and mitomycin C (MMC) is ascribed largely to their ability to generate interstrand crosslinks (ICLs) in DNA, which block the progression of replication forks. The processing of ICLs requires the Fanconi anemia (FA) pathway, excision repair, and translesion DNA synthesis (TLS). It also requires homologous recombination (HR), which repairs double-strand breaks (DSBs) generated by cleavage of the blocked replication forks. Here we describe KIAA1018, an evolutionarily conserved protein that has an N-terminal ubiquitin-binding zinc finger (UBZ) and a C-terminal nuclease domain. KIAA1018 is a 5'-->3' exonuclease and a structure-specific endonuclease that preferentially incises 5' flaps. Like cells from FA patients, human cells depleted of KIAA1018 are sensitized to ICL-inducing agents and display chromosomal instability. The link of KIAA1018 to the FA pathway is further strengthened by its recruitment to DNA damage through interaction of its UBZ domain with monoubiquitylated FANCD2. We therefore propose to name KIAA1018 FANCD2-associated nuclease, FAN1.
Collapse
Affiliation(s)
- Katja Kratz
- Institute of Molecular Cancer Research, University of Zurich, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Jowsey PA, Williams FM, Blain PG. The role of homologous recombination in the cellular response to sulphur mustard. Toxicol Lett 2010; 197:12-8. [DOI: 10.1016/j.toxlet.2010.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 04/21/2010] [Accepted: 04/23/2010] [Indexed: 01/19/2023]
|
158
|
Wang C, Lambert MW. The Fanconi anemia protein, FANCG, binds to the ERCC1-XPF endonuclease via its tetratricopeptide repeats and the central domain of ERCC1. Biochemistry 2010; 49:5560-9. [PMID: 20518486 DOI: 10.1021/bi100584c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is evidence that Fanconi anemia (FA) proteins play an important role in the repair of DNA interstrand cross-links (ICLs), but the precise mechanism by which this occurs is not clear. One of the critical steps in the ICL repair process involves unhooking of the cross-link from DNA by incisions on one strand on either side of the ICL and its subsequent removal. The ERCC1-XPF endonuclease is involved in this unhooking step and in the removal of the cross-link. We have previously shown that several of the FA proteins are needed to produce incisions created by ERCC1-XPF at sites of ICLs. To more clearly establish a link between FA proteins and the incision step(s) mediated by ERCC1-XPF, we undertook yeast two-hybrid analysis to determine whether FANCA, FANCC, FANCF, and FANCG directly interact with ERCC1 and XPF and, if so, to determine the sites of interaction. One of these FA proteins, FANCG, was found to have a strong affinity for ERCC1 and a moderate affinity for XPF. FANCG has been shown to contain seven tetratricopeptide repeat (TPR) motifs, which are motifs that mediate protein-protein interactions. Mapping the sites of interaction of FANCG with ERCC1, using site-directed mutagenesis, demonstrated that TPRs 1, 3, 5, and 6 are needed for binding of FANCG to ERCC1. ERCC1, in turn, was shown to interact with FANCG via its central domain, which is different from the region of ERCC1 that binds to XPF. This binding between FANCG and the ERCC1-XPF endonuclease, combined with our previous studies which show that FANCG is involved in the incision step mediated by ERCC1-XPF, establishes a link between an FA protein and the critical unhooking step of the ICL repair process.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Pathology and Laboratory Medicine, New Jersey Medical School and Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA
| | | |
Collapse
|
159
|
Issaeva N, Thomas HD, Djureinovic T, Djurenovic T, Jaspers JE, Stoimenov I, Kyle S, Pedley N, Gottipati P, Zur R, Sleeth K, Chatzakos V, Mulligan EA, Lundin C, Gubanova E, Kersbergen A, Harris AL, Sharma RA, Rottenberg S, Curtin NJ, Helleday T. 6-thioguanine selectively kills BRCA2-defective tumors and overcomes PARP inhibitor resistance. Cancer Res 2010; 70:6268-76. [PMID: 20631063 DOI: 10.1158/0008-5472.can-09-3416] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Familial breast and ovarian cancers are often defective in homologous recombination (HR) due to mutations in the BRCA1 or BRCA2 genes. Cisplatin chemotherapy or poly(ADP-ribose) polymerase (PARP) inhibitors were tested for these tumors in clinical trials. In a screen for novel drugs that selectively kill BRCA2-defective cells, we identified 6-thioguanine (6TG), which induces DNA double-strand breaks (DSB) that are repaired by HR. Furthermore, we show that 6TG is as efficient as a PARP inhibitor in selectively killing BRCA2-defective tumors in a xenograft model. Spontaneous BRCA1-defective mammary tumors gain resistance to PARP inhibitors through increased P-glycoprotein expression. Here, we show that 6TG efficiently kills such BRCA1-defective PARP inhibitor-resistant tumors. We also show that 6TG could kill cells and tumors that have gained resistance to PARP inhibitors or cisplatin through genetic reversion of the BRCA2 gene. Although HR is reactivated in PARP inhibitor-resistant BRCA2-defective cells, it is not fully restored for the repair of 6TG-induced lesions. This is likely to be due to several recombinogenic lesions being formed after 6TG. We show that BRCA2 is also required for survival from mismatch repair-independent lesions formed by 6TG, which do not include DSBs. This suggests that HR is involved in the repair of 6TG-induced DSBs as well as mismatch repair-independent 6TG-induced DNA lesion. Altogether, our data show that 6TG efficiently kills BRCA2-defective tumors and suggest that 6TG may be effective in the treatment of advanced tumors that have developed resistance to PARP inhibitors or platinum-based chemotherapy.
Collapse
Affiliation(s)
- Natalia Issaeva
- Department of Genetics, Microbiology, and Toxicology, Stockholm University, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Rahn JJ, Adair GM, Nairn RS. Multiple roles of ERCC1-XPF in mammalian interstrand crosslink repair. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:567-581. [PMID: 20658648 DOI: 10.1002/em.20583] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
DNA interstrand crosslinks (ICLs) are among the most deleterious cytotoxic lesions encountered by cells, mainly due to the covalent linkage these lesions create between the two strands of DNA which effectively blocks replication and transcription. Although ICL repair in mammalian cells is not fully understood, processing of these lesions is thought to begin by "unhooking" at the site of the damaged base accompanied by the generation of a double strand break and ultimately repair through translesion synthesis and homologous recombination. A key player in this repair process is the heterodimeric protein complex ERCC1-XPF. Although some models of ICL repair restrict ERCC1-XPF activity to the unhooking step, recent data suggest that this protein complex acts in additional downstream steps. Here, we review the evidence implicating ERCC1-XPF in multiple steps of ICL repair.
Collapse
Affiliation(s)
- Jennifer J Rahn
- Department of Carcinogenesis, Science Park-Research Division, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957, USA.
| | | | | |
Collapse
|
161
|
Vasquez KM. Targeting and processing of site-specific DNA interstrand crosslinks. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:527-39. [PMID: 20196133 PMCID: PMC2895014 DOI: 10.1002/em.20557] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
DNA interstrand crosslinks (ICLs) are among the most cytotoxic types of DNA damage, and thus ICL-inducing agents such as cyclophosphamide, melphalan, cisplatin, psoralen, and mitomycin C have been used clinically as anticancer drugs for decades. ICLs can also be formed endogenously as a consequence of cellular metabolic processes. ICL-inducing agents continue to be among the most effective chemotherapeutic treatments for many cancers; however, treatment with these agents can lead to secondary malignancies, in part due to mutagenic processing of the DNA lesions. The mechanisms of ICL repair have been characterized more thoroughly in bacteria and yeast than in mammalian cells. Thus, a better understanding of the molecular mechanisms of ICL processing offers the potential to improve the efficacy of these drugs in cancer therapy. In mammalian cells, it is thought that ICLs are repaired by the coordination of proteins from several pathways, including nucleotide excision repair (NER), base excision repair (BER), mismatch repair (MMR), homologous recombination (HR), translesion synthesis (TLS), and proteins involved in Fanconi anemia (FA). In this review, we focus on the potential functions of NER, MMR, and HR proteins in the repair of and response to ICLs in human cells and in mice. We will also discuss a unique approach, using psoralen covalently linked to triplex-forming oligonucleotides to direct ICLs to specific sites in the mammalian genome.
Collapse
Affiliation(s)
- Karen M Vasquez
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Science Park-Research Division, Smithville, Texas 78957, USA.
| |
Collapse
|
162
|
Hlavin EM, Smeaton MB, Miller PS. Initiation of DNA interstrand cross-link repair in mammalian cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:604-24. [PMID: 20658650 PMCID: PMC2911644 DOI: 10.1002/em.20559] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Interstrand cross-links (ICLs) are among the most cytotoxic DNA lesions to cells because they prevent the two DNA strands from separating, thereby precluding replication and transcription. Even though chemotherapeutic cross-linking agents are well established in clinical use, and numerous repair proteins have been implicated in the initial events of mammalian ICL repair, the precise mechanistic details of these events remain to be elucidated. This review will summarize our current understanding of how ICL repair is initiated with an emphasis on the context (replicating, transcribed or quiescent DNA) in which the ICL is recognized, and how the chemical and physical properties of ICLs influence repair. Although most studies have focused on replication-dependent repair because of the relation to highly replicative tumor cells, replication-independent ICL repair is likely to be important in the circumvention of cross-link cytotoxicity in nondividing, terminally differentiated cells that may be challenged with exogenous or endogenous sources of ICLs. Consequently, the ICL repair pathway that should be considered "dominant" appears to depend on the cell type and the DNA context in which the ICL is encountered. The ability to define and inhibit distinct pathways of ICL repair in different cell cycle phases may help in developing methods that increase cytotoxicity to cancer cells while reducing side-effects in nondividing normal cells. This may also lead to a better understanding of pathways that protect against malignancy and aging.
Collapse
Affiliation(s)
| | | | - Paul S. Miller
- Correspondence should be addressed to Paul S. Miller, , Phone: (410)-955-3489, Fax: (410)-955-2926
| |
Collapse
|
163
|
Legerski RJ. Repair of DNA interstrand cross-links during S phase of the mammalian cell cycle. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:540-551. [PMID: 20658646 PMCID: PMC2911997 DOI: 10.1002/em.20566] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
DNA interstrand cross-linking (ICL) agents are widely used in anticancer chemotherapy regimens, yet our understanding of the DNA repair mechanisms by which these lesions are removed from the genome remains incomplete. This is at least in part due to the enormously complicated nature and variety of the biochemical pathways that operate on these complex lesions. In this review, we have focused specifically on the S-phase pathway of ICL repair in mammalian cells, which appears to be the major mechanism by which these lesions are removed in cycling cells. The various stages and components of this pathway are discussed, and a putative molecular model is presented. In addition, we propose an explanation as to how this pathway can lead to the observed high levels of sister chromatid exchanges known to be induced by ICLs.
Collapse
Affiliation(s)
- Randy J Legerski
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| |
Collapse
|
164
|
Ho TV, Schärer OD. Translesion DNA synthesis polymerases in DNA interstrand crosslink repair. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:552-566. [PMID: 20658647 DOI: 10.1002/em.20573] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
DNA interstrand crosslinks (ICLs) are induced by a number of bifunctional antitumor drugs such as cisplatin, mitomycin C, or the nitrogen mustards as well as endogenous agents formed by lipid peroxidation. The repair of ICLs requires the coordinated interplay of a number of genome maintenance pathways, leading to the removal of ICLs through at least two distinct mechanisms. The major pathway of ICL repair is dependent on replication, homologous recombination, and the Fanconi anemia (FA) pathway, whereas a minor, G0/G1-specific and recombination-independent pathway depends on nucleotide excision repair. A central step in both pathways in vertebrates is translesion synthesis (TLS) and mutants in the TLS polymerases Rev1 and Pol zeta are exquisitely sensitive to crosslinking agents. Here, we review the involvement of Rev1 and Pol zeta as well as additional TLS polymerases, in particular, Pol eta, Pol kappa, Pol iota, and Pol nu, in ICL repair. Biochemical studies suggest that multiple TLS polymerases have the ability to bypass ICLs and that the extent ofbypass depends upon the structure as well as the extent of endo- or exonucleolytic processing of the ICL. As has been observed for lesions that affect only one strand of DNA, TLS polymerases are recruited by ubiquitinated proliferating nuclear antigen (PCNA) to repair ICLs in the G0/G1 pathway. By contrast, this data suggest that a different mechanism involving the FA pathway is operative in coordinating TLS in the context of replication-dependent ICL repair.
Collapse
Affiliation(s)
- The Vinh Ho
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York 11794-3400, USA
| | | |
Collapse
|
165
|
Wood RD. Mammalian nucleotide excision repair proteins and interstrand crosslink repair. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:520-6. [PMID: 20658645 PMCID: PMC3017513 DOI: 10.1002/em.20569] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Although various schemes for interstrand crosslink (ICL) repair incorporate DNA recombination, replication, and double-strand break intermediate steps, action of the nucleotide excision repair (NER) system or some variation of it is a common feature of most models. In the bacterium Escherichia coli, the NER enzyme UvrABC can incise on either side of an ICL to unhook the crosslink, and can proceed via a subsequent recombination step. The relevance of NER to ICL repair in mammalian cells has been challenged. Of all NER mutants, it is clear that ERCC1 and XPF-defective cells show the most pronounced sensitivities to ICL-inducing agents, and defects in ICL repair. However, there is good evidence that cells defective in NER proteins including XPA and XPG are also more sensitive than normal to ICL-inducing agents. These results are summarized here, together with evidence for defective crosslink removal in NER-defective cells. Studies of incision at sites of ICL by cell extracts and purified proteins have been done, but these studies are not all consistent with one another and further research is required.
Collapse
Affiliation(s)
- Richard D Wood
- Department of Carcinogenesis and The University of Texas Graduate School of Biomedical Sciences at Houston, The University of Texas M. D. Anderson Cancer Center, Science Park-Research Division, Smithville, Texas 78957, USA.
| |
Collapse
|
166
|
Cattell E, Sengerová B, McHugh PJ. The SNM1/Pso2 family of ICL repair nucleases: from yeast to man. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:635-645. [PMID: 20175117 DOI: 10.1002/em.20556] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Efficient interstrand crosslink (ICL) repair in yeast depends on the Pso2/Snm1 protein. Pso2 is a member of the highly conserved metallo-beta-lactamase structural family of nucleases. Mammalian cells possess three SNM1/Pso2 related proteins, SNM1A, SNM1B/Apollo, and SNM1C/Artemis. Evidence that SNM1A and SNM1B contribute to ICL repair is mounting, whereas Artemis appears to primarily contribute to non-ICL repair pathways, particularly some double-strand break repair events. Yeast Pso2 and all three mammalian SNM1-family proteins have been shown to possess nuclease activity. Here, we review the biochemical, genetic, and cellular evidence for the SNM1 family as DNA repair factors, focusing on ICL repair.
Collapse
Affiliation(s)
- Emma Cattell
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | | | | |
Collapse
|
167
|
Hinz JM. Role of homologous recombination in DNA interstrand crosslink repair. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:582-603. [PMID: 20658649 DOI: 10.1002/em.20577] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Homologous recombination repair (HRR) encompasses mechanisms that employ homologous DNA sequences as templates for repair or tolerance of a wide range of DNA lesions that inhibit DNA replication in S phase. Arguably the most imposing of these DNA lesions is that of the interstrand crosslink (ICL), consisting of a covalently attached chemical bridge between opposing DNA strands. ICL repair requires the coordinated activities of HRR and a number of proteins from other DNA repair and damage response systems, including nucleotide excision repair, base excision repair, mismatch repair, and translesion DNA synthesis (TLS). Interestingly, different organisms favor alternative methods of HRR in the ICL repair process. E. coli perform ICL repair using a homology-driven damage bypass mechanism analogous to daughter strand gap repair. Eukaryotes from yeast to humans initiate ICL repair primarily during DNA replication, relying on HRR activity to restart broken replication forks associated with double-strand break intermediates induced by nucleolytic activities of other excision repair factors. Higher eukaryotes also employ several additional factors, including members of the Fanconi anemia damage-response network, which further promote replication-associated ICL repair through the activation and coordination of various DNA excision repair, TLS, and HRR proteins. This review focuses on the proteins and general mechanisms of HRR associated with ICL repair in different model organisms.
Collapse
Affiliation(s)
- John M Hinz
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
168
|
McVey M. Strategies for DNA interstrand crosslink repair: insights from worms, flies, frogs, and slime molds. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:646-658. [PMID: 20143343 DOI: 10.1002/em.20551] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
DNA interstrand crosslinks (ICLs) are complex lesions that covalently link both strands of the DNA double helix and impede essential cellular processes such as DNA replication and transcription. Recent studies suggest that multiple repair pathways are involved in their removal. Elegant genetic analysis has demonstrated that at least three distinct sets of pathways cooperate in the repair and/or bypass of ICLs in budding yeast. Although the mechanisms of ICL repair in mammals appear similar to those in yeast, important differences have been documented. In addition, mammalian crosslink repair requires other repair factors, such as the Fanconi anemia proteins, whose functions are poorly understood. Because many of these proteins are conserved in simpler metazoans, nonmammalian models have become attractive systems for studying the function(s) of key crosslink repair factors. This review discusses the contributions that various model organisms have made to the field of ICL repair. Specifically, it highlights how studies performed with C. elegans, Drosophila, Xenopus, and the social amoeba Dictyostelium serve to complement those from bacteria, yeast, and mammals. Together, these investigations have revealed that although the underlying themes of ICL repair are largely conserved, the complement of DNA repair proteins utilized and the ways in which each of the proteins is used can vary substantially between different organisms.
Collapse
Affiliation(s)
- Mitch McVey
- Department of Biology, Tufts University, Medford, Massachusetts 02155, USA.
| |
Collapse
|
169
|
Yun J, Kim KM, Kim ST, Kim JH, Kim JA, Kong JH, Lee SH, Won YW, Sun JM, Lee J, Park SH, Park JO, Park YS, Lim HY, Kang WK. Predictive value of the ERCC1 expression for treatment response and survival in advanced gastric cancer patients receiving cisplatin-based first-line chemotherapy. Cancer Res Treat 2010; 42:101-6. [PMID: 20622964 DOI: 10.4143/crt.2010.42.2.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 01/20/2010] [Indexed: 01/06/2023] Open
Abstract
PURPOSE The aim of this study was to determine whether the ERCC1 expression is effective to predict the clinical outcomes of patients with advanced gastric cancer (AGC) and who were treated with cisplatin-based first-line chemotherapy. MATERIALS AND METHODS A total of 89 measurable AGC patients received cisplatin and capecitabine, with or without epirubicin, as a part of a randomized phase II study. Patients were included for the current molecular analysis if they had received two or more cycles of chemotherapy, their objective tumor responses were measured and if their paraffin-embedded tumor samples were available. The ERCC1 expression was examined by performing immunohistochemical (IHC) staining, and the patients were divided into two groups (positive or negative) according to the presence of IHC staining of the tumor cell nuclei. RESULTS Of the 32 eligible patients, 21 patients (66%) had tumor with a positive expression of ERCC1 and the remaining 11 patients had tumor with a negative ERCC1-expression. The ERCC1-negative patients achieved a higher response rate than that of the ERCC1-positive patients (44% vs. 28%, respectively), although the difference was not statistically significant (p=0.42). The median survival time for the all patients was 14.6 months (95% CI: 13.6 to 15.6 months). The one-year survival rate was similar for the ERCC1-negative patients (61%) and the ERCC1-positive patients (70%). CONCLUSION In the current study, the tumor ERCC1 expression by IHC staining could not predict the clinical response or survival of AGC patients who were treated with cisplatin-based first-line chemotherapy. The ERCC1 protein expression does not appear to be a useful tool for the selection of tailored chemotherapy for these patients.
Collapse
Affiliation(s)
- Jina Yun
- Department of Medicine, Sungkyunkwan University Samsung Medical Center, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Chen S, Zhang J, Wang R, Luo X, Chen H. The platinum-based treatments for advanced non-small cell lung cancer, is low/negative ERCC1 expression better than high/positive ERCC1 expression? A meta-analysis. Lung Cancer 2010; 70:63-70. [PMID: 20541281 DOI: 10.1016/j.lungcan.2010.05.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 05/01/2010] [Accepted: 05/07/2010] [Indexed: 12/15/2022]
Abstract
BACKGROUND The predictive value of ERCC1 for prognosis and sensitivity to platinum-based chemotherapy in patients with non-small cell lung cancer (NSCLC) remains controversial. This meta-analysis was performed to provide an assessment of whether expression variations of ERCC1 are associated with objective response and median survival in patients with advanced NSCLC treated with platinum-based chemotherapy. METHODS We searched MEDLINE, EMBASE and CNKI for all eligible studies and conducted a meta-analysis of 12 studies (n=836 patients) that evaluated the correlation between ERCC1 levels (detected by immunohistochemistry or real-time reverse transcriptase PCR) and objective response or median survival in patients receiving platinum-based chemotherapy for advanced NSCLC. Pooled odds ratios (OR) for the objective response were calculated using the Mantel-Haenszel method. Pooled median ratios for median survival were calculated using the weighted sum of the log-ratio of median ratios from each individual study. RESULTS Among 836 tumors, ERCC1 expression was high/positive in 416 (49.8%) and low/negative in 420 (50.2%). Response to platinum-based chemotherapy was significantly higher in patients with ERCC1 low/negative expression (OR=0.48; 95% CI, 0.35-0.64; P<0.00001). Median survival time was significantly prolonged when ERCC1 low/negative expression was compared with ERCC1 high/positive expression (MR: 0.77; 95% CI, 0.47-1.07; P<0.00001). CONCLUSIONS Low/negative expression of ERCC1 was associated with higher objective response and median survival in advanced NSCLC patients treated with platinum-based chemotherapy. ERCC1 may be a suitable marker of prognosis and sensitivity to platinum-based chemotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Sufeng Chen
- Department of Cardiothoracic Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai 200233, People's Republic of China
| | | | | | | | | |
Collapse
|
171
|
Zhang J, Gu SY, Zhang P, Jia Z, Chang JH. ERCC2 Lys751Gln polymorphism is associated with lung cancer among Caucasians. Eur J Cancer 2010; 46:2479-84. [PMID: 20627704 DOI: 10.1016/j.ejca.2010.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Revised: 04/28/2010] [Accepted: 05/04/2010] [Indexed: 10/19/2022]
Abstract
To derive a more precise estimation of the relationship between the excision repair cross-complementing rodent repair deficiency, group 2 (ERCC2) Lys751Gln polymorphism and lung cancer risk, a meta-analysis was performed. A total of 23 studies including 8137 cases and 9824 controls were involved in this meta-analysis. Overall, significantly elevated lung cancer risk was associated with ERCC2 Gln allele when all studies were pooled into the meta-analysis (Lys/Gln versus Lys/Lys: odds ratio (OR)=1.10, 95% confidence interval (CI)=1.03-1.19; Gln/Gln versus Lys/Lys: OR=1.20, 95% CI=1.06-1.35; dominant model: OR=1.13, 95% CI=1.05-1.20; and recessive model: OR=1.15, 95% CI=1.03-1.29). In the subgroup analysis by ethnicity, significantly increased risk was only found for Caucasians (Gln/Gln versus Lys/Lys: OR=1.25, 95% CI=1.08-1.45; dominant model: OR=1.10, 95% CI=1.00-1.22; and recessive model: OR=1.22, 95% CI=1.06-1.40). When stratified by study design, statistically significantly elevated risks were found in hospital-based studies (Lys/Gln versus Lys/Lys: OR=1.12, 95% CI=1.03-1.22; Gln/Gln versus Lys/Lys: OR=1.24, 95% CI=1.06-1.44; dominant model: OR=1.15, 95% CI=1.06-1.24; and recessive model: OR=1.19, 95% CI=1.03-1.37) and population-based studies (Gln/Gln versus Lys/Lys: OR=1.57, 95% CI=1.12-2.20 and recessive model: OR=1.50, 95% CI=1.08-2.07). In the subgroup analysis whether or not the studies were matched on smoking, significantly increased risk was found not in those matched studies but in the unmatched studies (Lys/Gln versus Lys/Lys: OR=1.11, 95% CI=1.03-1.19; Gln/Gln versus Lys/Lys: OR=1.22, 95% CI=1.07-1.40; dominant model: OR=1.13, 95% CI=1.05-1.22; and recessive model: OR=1.18, 95% CI=1.04-1.33). In conclusion, this meta-analysis suggests that the ERCC2 Lys751Gln polymorphism may contribute to lung cancer susceptibility among Caucasians.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Medical Oncology, Cancer Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
172
|
Chan SH, Cheung FMF, Ng WT, Choi CW, Cheung KN, Yiu KH, Lee AWM. Can the analysis of ERCC1 expression contribute to individualized therapy in nasopharyngeal carcinoma? Int J Radiat Oncol Biol Phys 2010; 79:1414-20. [PMID: 20605357 DOI: 10.1016/j.ijrobp.2009.12.072] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 12/18/2009] [Accepted: 12/21/2009] [Indexed: 10/19/2022]
Abstract
PURPOSE To analyze the expression of excision repair cross-complementation group 1 (ERCC1) protein in predicting the clinical outcome of nasopharyngeal carcinoma (NPC). METHODS AND MATERIALS The histologic specimens of 258 patients with Stage III to IVB nonkeratinizing NPC who were treated with radiotherapy alone (Group I) or concurrent-adjuvant chemoradiotherapy (Group II) were retrieved. Immunostaining on ERCC1 protein was performed. The relationship of ERCC1 expression and clinical outcomes was analyzed. RESULTS The median ERCC1 score (proportion score of positively stained cells times intensity) was 200 (range, 0-300), and ERCC1 expression was defined as high if the score was above the median. In Group I high-score tumor had a statistically lower locoregional failure-free rate (LRFFR) compared with low-score tumor (p < 0.05) but not distant failure-free rate (DFFR) and overall survival (OS). In Group II no statistically differences were noted in LRFFR, DFFR and OS with regard to the ERCC1 expression. Resistance to cisplatin-containing chemotherapy in high-ERCC1 score tumor was not observed in Group II. Interestingly, low-score tumor in Group I achieved similar local and distant control compared with Group II. Multivariate analysis showed that ERCC1 score was an independent prognostic factor in LRFFR (p < 0.05) and approached statistical significance in failure-free survival (p = 0.08) and OS (p = 0.07). Tumor with high ERCC1 score had a 2-fold (95% confidence interval, 1.02-3.85) increased risk of locoregional failure. This may imply an association of ERCC1 expression with the repair of radiation damage. CONCLUSIONS High ERCC1 expression predicts poor locoregional control in NPC. Chemotherapy response is not affected by ERCC1 expression. Further validation is required.
Collapse
Affiliation(s)
- Siu Hong Chan
- Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong.
| | | | | | | | | | | | | |
Collapse
|
173
|
Zhang J, Qiu LX, Leaw SJ, Hu XC, Chang JH. The association between XPD Asp312Asn polymorphism and lung cancer risk: a meta-analysis including 16,949 subjects. Med Oncol 2010; 28:655-60. [PMID: 20354818 DOI: 10.1007/s12032-010-9501-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 03/11/2010] [Indexed: 11/29/2022]
Abstract
To derive a more precise estimation of the relationship between the xeroderma pigmentosum group D (XPD) Asp312Asn polymorphism and lung cancer risk, a meta-analysis was performed. PubMed, Medline, Embase, and Web of Science were searched. Crude ORs with 95% CIs were used to assess the strength of association between the XPD Asp312Asn polymorphism and lung cancer risk. The pooled ORs were performed with co-dominant model (Asp/Asn vs. Asp/Asp, Asn/Asn vs. Asp/Asp), dominant model (Asp/Asn + Asn/Asn vs. Asp/Asp), and recessive model (Asn/Asn vs. Asp/Asp+Asp/Asn), respectively. A total of 18 studies including 7,552 cases and 9,397 controls were involved in this meta-analysis. Overall, significantly elevated lung cancer risk was associated with XPD Asn allele when all studies were pooled into the meta-analysis (Asn/Asn vs. Asp/Asp: OR=1.158, 95% CI=1.018-1.317; recessive model: OR=1.161, 95% CI=1.029-1.311). In the subgroup analysis by ethnicity, significantly increased risks were found for both Caucasians (Asn/Asn vs. Asp/Asp: OR=1.164, 95% CI=1.003-1.351; recessive model: OR=1.169, 95% CI=1.016-1.345) and Asians (Asn/Asn vs. Asp/Asp: OR=8.056, 95% CI=2.420-26.817; recessive model: OR=7.956, 95% CI=2.391-26.477). When stratified by study design, statistically significantly elevated risk was noted in hospital-based studies (Asn/Asn vs. Asp/Asp: OR=1.315, 95% CI=1.110-1.558; recessive model: OR=1.290, 95% CI=1.099-1.513). In conclusion, this meta-analysis suggests that the XPD Asn allele is a low-penetrant risk factor for developing lung cancer.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Medical Oncology, Cancer Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
174
|
Abstract
Homologous recombination (HR) performs crucial functions including DNA repair, segregation of homologous chromosomes, propagation of genetic diversity, and maintenance of telomeres. HR is responsible for the repair of DNA double-strand breaks and DNA interstrand cross-links. The process of HR is initiated at the site of DNA breaks and gaps and involves a search for homologous sequences promoted by Rad51 and auxiliary proteins followed by the subsequent invasion of broken DNA ends into the homologous duplex DNA that then serves as a template for repair. The invasion produces a cross-stranded structure, known as the Holliday junction. Here, we describe the properties of Rad54, an important and versatile HR protein that is evolutionarily conserved in eukaryotes. Rad54 is a motor protein that translocates along dsDNA and performs several important functions in HR. The current review focuses on the recently identified Rad54 activities which contribute to the late phase of HR, especially the branch migration of Holliday junctions.
Collapse
Affiliation(s)
- Alexander V Mazin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| | | | | | | |
Collapse
|
175
|
Muniandy PA, Liu J, Majumdar A, Liu ST, Seidman MM. DNA interstrand crosslink repair in mammalian cells: step by step. Crit Rev Biochem Mol Biol 2010; 45:23-49. [PMID: 20039786 PMCID: PMC2824768 DOI: 10.3109/10409230903501819] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Interstrand DNA crosslinks (ICLs) are formed by natural products of metabolism and by chemotherapeutic reagents. Work in E. coli identified a two cycle repair scheme involving incisions on one strand on either side of the ICL (unhooking) producing a gapped intermediate with the incised oligonucleotide attached to the intact strand. The gap is filled by recombinational repair or lesion bypass synthesis. The remaining monoadduct is then removed by nucleotide excision repair (NER). Despite considerable effort, our understanding of each step in mammalian cells is still quite limited. In part this reflects the variety of crosslinking compounds, each with distinct structural features, used by different investigators. Also, multiple repair pathways are involved, variably operative during the cell cycle. G(1) phase repair requires functions from NER, although the mechanism of recognition has not been determined. Repair can be initiated by encounters with the transcriptional apparatus, or a replication fork. In the case of the latter, the reconstruction of a replication fork, stalled or broken by collision with an ICL, adds to the complexity of the repair process. The enzymology of unhooking, the identity of the lesion bypass polymerases required to fill the first repair gap, and the functions involved in the second repair cycle are all subjects of active inquiry. Here we will review current understanding of each step in ICL repair in mammalian cells.
Collapse
Affiliation(s)
- Parameswary A Muniandy
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
176
|
Effect of the anti-neoplastic drug doxorubicin on XPD-mutated DNA repair-deficient human cells. DNA Repair (Amst) 2010; 9:40-7. [DOI: 10.1016/j.dnarep.2009.10.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 10/07/2009] [Accepted: 10/08/2009] [Indexed: 11/22/2022]
|
177
|
Differential roles for DNA polymerases eta, zeta, and REV1 in lesion bypass of intrastrand versus interstrand DNA cross-links. Mol Cell Biol 2009; 30:1217-30. [PMID: 20028736 DOI: 10.1128/mcb.00993-09] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
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.
Collapse
|
178
|
Zietlow L, Smith LA, Bessho M, Bessho T. Evidence for the involvement of human DNA polymerase N in the repair of DNA interstrand cross-links. Biochemistry 2009; 48:11817-24. [PMID: 19908865 PMCID: PMC2790558 DOI: 10.1021/bi9015346] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human DNA polymerase N (PolN) is an A-family nuclear DNA polymerase whose function is unknown. This study examines the possible role of PolN in DNA repair in human cells treated with PolN-targeted siRNA. HeLa cells with siRNA-mediated knockdown of PolN were more sensitive than control cells to DNA cross-linking agent mitomycin C (MMC) but were not hypersensitive to UV irradiation. The MMC hypersensitivity of PolN knockdown cells was rescued by the overexpression of DNA polymerase-proficient PolN but not by DNA polymerase-deficient PolN. Furthermore, in vitro experiments showed that purified PolN conducts low-efficiency nonmutagenic bypass of a psoralen DNA interstrand cross-link (ICL), whose structure resembles an intermediate in the proposed pathway of ICL repair. These results suggest that PolN might play a role in translesion DNA synthesis during ICL repair in human cells.
Collapse
Affiliation(s)
- Laura Zietlow
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, U.S.A
| | - Leigh Anne Smith
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, U.S.A
| | - Mika Bessho
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, U.S.A
| | - Tadayoshi Bessho
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, U.S.A
| |
Collapse
|
179
|
Yin Z, Su M, Li X, Li M, Ma R, He Q, Zhou B. ERCC2, ERCC1 polymorphisms and haplotypes, cooking oil fume and lung adenocarcinoma risk in Chinese non-smoking females. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2009; 28:153. [PMID: 20003391 PMCID: PMC2797795 DOI: 10.1186/1756-9966-28-153] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 12/14/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND Excision repair cross-complementing group 1 (ERCC1) and group 2 (ERCC2) proteins play important roles in the repair of DNA damage and adducts. Single nucleotide polymorphisms (SNPs) of DNA repair genes are suspected to influence the risk of lung cancer. This study aimed to investigate the association between the ERCC2 751, 312 and ERCC1 118 polymorphisms and the risk of lung adenocarcinoma in Chinese non-smoking females. METHODS A hospital-based case-control study of 285 patients and 285 matched controls was conducted. Information concerning demographic and risk factors was obtained for each case and control by a trained interviewer. After informed consent was obtained, each person donated 10 ml blood for biomarker testing. Three polymorphisms were determined by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. RESULTS This study showed that the individuals with the combined ERCC2 751AC/CC genotypes were at an increased risk for lung adenocarcinoma compared with those carrying the AA genotype [adjusted odds ratios (OR) 1.64, 95% confidence interval (CI) 1.06-2.52]. The stratified analysis suggested that increased risk associated with ERCC2 751 variant genotypes (AC/CC) was more pronounced in individuals without exposure to cooking oil fume (OR 1.98, 95%CI 1.18-3.32) and those without exposure to fuel smoke (OR 2.47, 95%CI 1.46-4.18). Haplotype analysis showed that the A-G-T and C-G-C haplotypes were associated with increased risk of lung adenocarcinoma among non-smoking females (ORs were 1.43 and 2.28, 95%CIs were 1.07-1.91 and 1.34-3.89, respectively). CONCLUSION ERCC2 751 polymorphism may be a genetic risk modifier for lung adenocarcinoma in non-smoking females in China.
Collapse
Affiliation(s)
- Zhihua Yin
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang 110001, PR China.
| | | | | | | | | | | | | |
Collapse
|
180
|
Abstract
Fanconi Anemia (FA) is an inherited genomic instability disorder, caused by mutations in genes regulating replication-dependent removal of interstrand DNA crosslinks. The Fanconi Anemia pathway is thought to coordinate a complex mechanism that enlists elements of three classic DNA repair pathways, namely homologous recombination, nucleotide excision repair, and mutagenic translesion synthesis, in response to genotoxic insults. To this end, the Fanconi Anemia pathway employs a unique nuclear protein complex that ubiquitinates FANCD2 and FANCI, leading to formation of DNA repair structures. Lack of obvious enzymatic activities among most FA members has made it challenging to unravel its precise modus operandi. Here we review the current understanding of how the Fanconi Anemia pathway components participate in DNA repair and discuss the mechanisms that regulate this pathway to ensure timely, efficient, and correct restoration of chromosomal integrity.
Collapse
Affiliation(s)
- George-Lucian Moldovan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
181
|
Bhagwat N, Olsen AL, Wang AT, Hanada K, Stuckert P, Kanaar R, D'Andrea A, Niedernhofer LJ, McHugh PJ. XPF-ERCC1 participates in the Fanconi anemia pathway of cross-link repair. Mol Cell Biol 2009; 29:6427-37. [PMID: 19805513 PMCID: PMC2786876 DOI: 10.1128/mcb.00086-09] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/28/2009] [Accepted: 09/26/2009] [Indexed: 11/20/2022] Open
Abstract
Interstrand cross-links (ICLs) prevent DNA strand separation and, therefore, transcription and replication, making them extremely cytotoxic. The precise mechanism by which ICLs are removed from mammalian genomes largely remains elusive. Genetic evidence implicates ATR, the Fanconi anemia proteins, proteins required for homologous recombination, translesion synthesis, and at least two endonucleases, MUS81-EME1 and XPF-ERCC1. ICLs cause replication-dependent DNA double-strand breaks (DSBs), and MUS81-EME1 facilitates DSB formation. The subsequent repair of these DSBs occurs via homologous recombination after the ICL is unhooked by XPF-ERCC1. Here, we examined the effect of the loss of either nuclease on FANCD2 monoubiquitination to determine if the nucleolytic processing of ICLs is required for the activation of the Fanconi anemia pathway. FANCD2 was monoubiquitinated in Mus81(-/-), Ercc1(-/-), and XPF-deficient human, mouse, and hamster cells exposed to cross-linking agents. However, the monoubiquitinated form of FANCD2 persisted longer in XPF-ERCC1-deficient cells than in wild-type cells. Moreover, the levels of chromatin-bound FANCD2 were dramatically reduced and the number of ICL-induced FANCD2 foci significantly lower in XPF-ERCC1-deficient cells. These data demonstrate that the unhooking of an ICL by XPF-ERCC1 is necessary for the stable localization of FANCD2 to the chromatin and subsequent homologous recombination-mediated DSB repair.
Collapse
Affiliation(s)
- Nikhil Bhagwat
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Anna L. Olsen
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Anderson T. Wang
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Katsuhiro Hanada
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Patricia Stuckert
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Roland Kanaar
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Alan D'Andrea
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Laura J. Niedernhofer
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| | - Peter J. McHugh
- Department of Human Genetics, University of Pittsburgh School of Public Health, A300 Crabtree Hall, 130 Desoto St., Pittsburgh, Pennsylvania 15261, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion 2.6, 5117 Centre Ave., Pittsburgh, Pennsylvania 15213-1863, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom, Department of Cell Biology & Genetics, Cancer Genomics Center, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St., Boston, Massachusetts 02115, Department of Radiation Oncology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, E1240 BSTWR, 200 Lothrop St., Pittsburgh, Pennsylvania 15261
| |
Collapse
|
182
|
Bhagwat NR, Roginskaya VY, Acquafondata MB, Dhir R, Wood RD, Niedernhofer LJ. Immunodetection of DNA repair endonuclease ERCC1-XPF in human tissue. Cancer Res 2009; 69:6831-8. [PMID: 19723666 DOI: 10.1158/0008-5472.can-09-1237] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The high incidence of resistance to DNA-damaging chemotherapeutic drugs and severe side effects of chemotherapy have led to a search for biomarkers able to predict which patients are most likely to respond to therapy. ERCC1-XPF nuclease is required for nucleotide excision repair of helix-distorting DNA damage and the repair of DNA interstrand crosslinks. Thus, it is essential for several pathways of repair of DNA damage by cisplatin and related drugs, which are widely used in the treatment of non-small cell lung carcinoma and other late-stage tumors. Consequently, there is tremendous interest in measuring ERCC1-XPF expression in tumor samples. Many immunohistochemistry studies have been done, but the antibodies for ERCC1-XPF were not rigorously tested for antigen specificity. Herein, we survey a battery of antibodies raised against human ERCC1 or XPF for their specificity using ERCC1-XPF-deficient cells as a negative control. Antibodies were tested for the following applications: immunoblotting, immunoprecipitation from cell extracts, immunofluorescence detection in fixed cells, colocalization of ERCC1-XPF with UV radiation-induced DNA damage in fixed cells, and immunohistochemistry in paraffin-embedded samples. Although several commercially available antibodies are suitable for immunodetection of ERCC1-XPF in some applications, only a select subset is appropriate for detection of this repair complex in fixed specimens. The most commonly used antibody, 8F1, is not suitable for immunodetection in tissue. The results with validated antibodies reveal marked differences in ERCC1-XPF protein levels between samples and cell types.
Collapse
Affiliation(s)
- Nikhil R Bhagwat
- Department of Human Genetics, University of Pittsburgh School of Public Health, PA, USA
| | | | | | | | | | | |
Collapse
|
183
|
Gari K, Constantinou A. The role of the Fanconi anemia network in the response to DNA replication stress. Crit Rev Biochem Mol Biol 2009; 44:292-325. [PMID: 19728769 DOI: 10.1080/10409230903154150] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Fanconi anemia is a genetically heterogeneous disorder associated with chromosome instability and a highly elevated risk for developing cancer. The mutated genes encode proteins involved in the cellular response to DNA replication stress. Fanconi anemia proteins are extensively connected with DNA caretaker proteins, and appear to function as a hub for the coordination of DNA repair with DNA replication and cell cycle progression. At a molecular level, however, the raison d'être of Fanconi anemia proteins still remains largely elusive. The thirteen Fanconi anemia proteins identified to date have not been embraced into a single and defined biological process. To help put the Fanconi anemia puzzle into perspective, we begin this review with a summary of the strategies employed by prokaryotes and eukaryotes to tolerate obstacles to the progression of replication forks. We then summarize what we know about Fanconi anemia with an emphasis on biochemical aspects, and discuss how the Fanconi anemia network, a late acquisition in evolution, may function to permit the faithful and complete duplication of our very large vertebrate chromosomes.
Collapse
Affiliation(s)
- Kerstin Gari
- DNA Damage Response Laboratory, Cancer Research UK, London Research Institute, Clare Hall Laboratories, South Mimms, UK
| | | |
Collapse
|
184
|
Smeaton MB, Hlavin EM, Noronha AM, Murphy SP, Wilds CJ, Miller PS. Effect of cross-link structure on DNA interstrand cross-link repair synthesis. Chem Res Toxicol 2009; 22:1285-97. [PMID: 19580249 DOI: 10.1021/tx9000896] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
DNA interstrand cross-links (ICLs) are products of chemotherapeutic agents and cellular metabolic processes that block both replication and transcription. If left unrepaired, ICLs are extremely toxic to cells, and ICL repair mechanisms contribute to the survival of certain chemotherapeutic resistance tumors. A critical step in ICL repair involves unhooking the cross-link. In the absence of a homologous donor sequence, the resulting gap can be filled in by a repair synthesis step involving bypass of the cross-link remnant. Here, we examine the effect of cross-link structure on the ability of unhooked DNA substrates to undergo repair synthesis in mammalian whole cell extracts. Using 32P incorporation assays, we found that repair synthesis occurs efficiently past the site of damage when a DNA substrate containing a single N4C-ethyl-N4C cross-link is incubated in HeLa or Chinese hamster ovary cell extracts. This lesion, which can base pair with deoxyguanosine, is readily bypassed by both Escherichia coli DNA polymerase I and T7 DNA polymerase in a primer extension assay. In contrast, bypass was not observed in the primer extension assay or in mammalian cell extracts when DNA substrates containing a N3T-ethyl-N3T or N1I-ethyl-N3T cross-link, whose linkers obstruct the hydrogen bond face of the bases, were used. A modified phosphorothioate sequencing method was used to analyze the ICL repair patches created in the mammalian cell extracts. In the case of the N4C-ethyl-N4C substrate, the repair patch spanned the site of the cross-link, and the lesion was bypassed in an error-free manner. However, although the N3T-ethyl-N3T and N1I-ethyl-N3T substrates were unhooked in the extracts, bypass was not detected. These and our previous results suggest that although the chemical structure of an ICL may not affect initial cross-link unhooking, it can play a significant role in subsequent processing of the cross-link. Understanding how the physical and chemical differences of ICLs affect repair may provide a better understanding of the cytotoxic and mutagenic potential of specific ICLs.
Collapse
Affiliation(s)
- Michael B Smeaton
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
| | | | | | | | | | | |
Collapse
|
185
|
Roh DS, Funderburgh JL. Impact on the corneal endothelium of mitomycin C during photorefractive keratectomy. J Refract Surg 2009; 25:894-7. [PMID: 19835330 DOI: 10.3928/1081597x-20090617-10] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 05/05/2009] [Indexed: 11/20/2022]
Abstract
PURPOSE This brief review examines both basic science and clinical studies to evaluate the potential impact on the health of the corneal endothelium of mitomycin C (MMC) usage during photorefractive keratectomy (PRK). METHODS The mechanism of action and consequences of MMC are reviewed within the context of in vitro, animal, and clinical studies and a hypothesis of how this vital cell layer responds to MMC at both the cellular and clinical levels is formed. RESULTS Seven basic science studies were reviewed demonstrating significant MMC toxicity to corneal endothelial cells. Of the five clinical studies reviewed, three demonstrated no effect on corneal endothelial density, whereas two studies found significant cell loss after MMC usage. CONCLUSIONS Although all of the basic science studies reviewed highlight the toxicity of MMC on the corneal endothelium, current clinical studies are less conclusive. Given the corneal penetration of MMC and the fragile nature of the corneal endothelium, additional follow-up studies are needed to determine the long-term impact of MMC usage during PRK on the corneal endothelium.
Collapse
Affiliation(s)
- Danny S Roh
- Department of Ophthalmology, UPMC Eye and Ear Institute, University of Pittsburgh School of Medicine, PA, USA
| | | |
Collapse
|
186
|
Wilson WR, Stribbling SM, Pruijn FB, Syddall SP, Patterson AV, Liyanage HS, Smith E, Botting KJ, Tercel M. Nitro-chloromethylbenzindolines: hypoxia-activated prodrugs of potent adenine N3 DNA minor groove alkylators. Mol Cancer Ther 2009; 8:2903-13. [DOI: 10.1158/1535-7163.mct-09-0571] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
187
|
Al-Minawi AZ, Lee YF, Håkansson D, Johansson F, Lundin C, Saleh-Gohari N, Schultz N, Jenssen D, Bryant HE, Meuth M, Hinz JM, Helleday T. The ERCC1/XPF endonuclease is required for completion of homologous recombination at DNA replication forks stalled by inter-strand cross-links. Nucleic Acids Res 2009; 37:6400-13. [PMID: 19713438 PMCID: PMC2770670 DOI: 10.1093/nar/gkp705] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 08/06/2009] [Accepted: 08/09/2009] [Indexed: 12/27/2022] Open
Abstract
Both the ERCC1-XPF complex and the proteins involved in homoIogous recombination (HR) have critical roles in inter-strand cross-link (ICL) repair. Here, we report that mitomycin C-induced lesions inhibit replication fork elongation. Furthermore, mitomycin C-induced DNA double-strand breaks (DSBs) are the result of the collapse of ICL-stalled replication forks. These are not formed through replication run off, as we show that mitomycin C or cisplatin-induced DNA lesions are not incised by global genome nucleotide excision repair (GGR). We also suggest that ICL-lesion repair is initiated either by replication or transcription, as the GGR does not incise ICL-lesions. Furthermore, we report that RAD51 foci are induced by cisplatin or mitomycin C independently of ERCC1, but that mitomycin C-induced HR measured in a reporter construct is impaired in ERCC1-defective cells. These data suggest that ERCC1-XPF plays a role in completion of HR in ICL repair. We also find no additional sensitivity to cisplatin by siRNA co-depletion of XRCC3 and ERCC1, showing that the two proteins act on the same pathway to promote survival.
Collapse
Affiliation(s)
- Ali Z. Al-Minawi
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Yin-Fai Lee
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Daniel Håkansson
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Fredrik Johansson
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Cecilia Lundin
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Nasrollah Saleh-Gohari
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Niklas Schultz
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Dag Jenssen
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Helen E. Bryant
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Mark Meuth
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - John M. Hinz
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| | - Thomas Helleday
- The Institute for Cancer Studies, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK, Department of Genetics Microbiology and Toxicology, Arrhenius Laboratory, Stockholm University, S-106 91 Stockholm, Sweden, Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, OX3 7DQ, UK and Genetic Department, Kerman University of Medical Sciences, Medical school, Bozorgrah Emam, Kerman, 76169-14111, Iran and School of Molecular Biosciences, Washington State University, Pullman, WA, 99164, USA
| |
Collapse
|
188
|
Abstract
DNA damage by agents crosslinking the strands presents a formidable challenge to the cell to repair for survival and to repair accurately for maintenance of genetic information. It appears that repair of DNA crosslinks occurs in a path involving double strand breaks (DSBs) in the DNA. Mammalian cells have multiple systems involved in the repair response to such damage, including the Fanconi anemia pathway that appears to be directly involved, although the mechanisms and site of action remain elusive. A particular finding relating to deficiency of the Fanconi anemia pathway is the observation of chromosomal radial formations after ICL damage. The basis of formation of such chromosomal aberrations is unknown although they appear secondarily to DSBs. Here we review the processes involved in response to DNA interstrand crosslinks which might lead to radial formation and the role of the nucleotide excision repair gene, ERCC1, which is required for a normal response, not just to DNA crosslinks, but also for DSBs at collapsed replication forks caused by substrate depletion.
Collapse
Affiliation(s)
- Kevin M. McCabe
- Department of Civil, Architectural, and Environmental Engineering, University of Colorado Boulder, Boulder, CO 80309
| | - Susan B. Olson
- Department of Molecular and Medical Genetics, OHSU, Sam Jackson Park Road, Portland, OR 97239
| | - Robb E. Moses
- Department of Molecular and Medical Genetics, OHSU, Sam Jackson Park Road, Portland, OR 97239
| |
Collapse
|
189
|
Roques C, Coulombe Y, Delannoy M, Vignard J, Grossi S, Brodeur I, Rodrigue A, Gautier J, Stasiak AZ, Stasiak A, Constantinou A, Masson JY. MRE11-RAD50-NBS1 is a critical regulator of FANCD2 stability and function during DNA double-strand break repair. EMBO J 2009; 28:2400-13. [PMID: 19609304 PMCID: PMC2735166 DOI: 10.1038/emboj.2009.193] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 06/17/2009] [Indexed: 01/04/2023] Open
Abstract
Monoubiquitination of the Fanconi anaemia protein FANCD2 is a key event leading to repair of interstrand cross-links. It was reported earlier that FANCD2 co-localizes with NBS1. However, the functional connection between FANCD2 and MRE11 is poorly understood. In this study, we show that inhibition of MRE11, NBS1 or RAD50 leads to a destabilization of FANCD2. FANCD2 accumulated from mid-S to G2 phase within sites containing single-stranded DNA (ssDNA) intermediates, or at sites of DNA damage, such as those created by restriction endonucleases and laser irradiation. Purified FANCD2, a ring-like particle by electron microscopy, preferentially bound ssDNA over various DNA substrates. Inhibition of MRE11 nuclease activity by Mirin decreased the number of FANCD2 foci formed in vivo. We propose that FANCD2 binds to ssDNA arising from MRE11-processed DNA double-strand breaks. Our data establish MRN as a crucial regulator of FANCD2 stability and function in the DNA damage response.
Collapse
Affiliation(s)
- Céline Roques
- Genome Stability Laboratory, Laval University Cancer Research Center, Hôtel-Dieu de Québec, Québec, Canada
| | - Yan Coulombe
- Genome Stability Laboratory, Laval University Cancer Research Center, Hôtel-Dieu de Québec, Québec, Canada
| | - Mathieu Delannoy
- Department of Biochemistry, University of Lausanne, BIL Biomedical Research Center, Lausanne, Switzerland
| | - Julien Vignard
- Genome Stability Laboratory, Laval University Cancer Research Center, Hôtel-Dieu de Québec, Québec, Canada
| | - Simona Grossi
- Department of Biochemistry, University of Lausanne, BIL Biomedical Research Center, Lausanne, Switzerland
| | - Isabelle Brodeur
- Genome Stability Laboratory, Laval University Cancer Research Center, Hôtel-Dieu de Québec, Québec, Canada
| | - Amélie Rodrigue
- Genome Stability Laboratory, Laval University Cancer Research Center, Hôtel-Dieu de Québec, Québec, Canada
| | - Jean Gautier
- Columbia University, Institute for Cancer Genetics, Irving Cancer Research Center, New York, NY, USA
| | - Alicja Z Stasiak
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Andrzej Stasiak
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Angelos Constantinou
- Department of Biochemistry, University of Lausanne, BIL Biomedical Research Center, Lausanne, Switzerland
| | - Jean-Yves Masson
- Genome Stability Laboratory, Laval University Cancer Research Center, Hôtel-Dieu de Québec, Québec, Canada
| |
Collapse
|
190
|
Muniandy PA, Thapa D, Thazhathveetil AK, Liu ST, Seidman MM. Repair of laser-localized DNA interstrand cross-links in G1 phase mammalian cells. J Biol Chem 2009; 284:27908-27917. [PMID: 19684342 DOI: 10.1074/jbc.m109.029025] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interstrand cross-links (ICLs) are absolute blocks to transcription and replication and can provoke genomic instability and cell death. Studies in bacteria define a two-stage repair scheme, the first involving recognition and incision on either side of the cross-link on one strand (unhooking), followed by recombinational repair or lesion bypass synthesis. The resultant monoadduct is removed in a second stage by nucleotide excision repair. In mammalian cells, there are multiple, but poorly defined, pathways, with much current attention on repair in S phase. However, many questions remain, including the efficiency of repair in the absence of replication, the factors involved in cross-link recognition, and the timing and demarcation of the first and second repair cycles. We have followed the repair of laser-localized lesions formed by psoralen (cross-links/monoadducts) and angelicin (only monoadducts) in mammalian cells. Both were repaired in G(1) phase by nucleotide excision repair-dependent pathways. Removal of psoralen adducts was blocked in XPC-deficient cells but occurred with wild type kinetics in cells deficient in DDB2 protein (XPE). XPC protein was rapidly recruited to psoralen adducts. However, accumulation of DDB2 was slow and XPC-dependent. Inhibition of repair DNA synthesis did not interfere with DDB2 recruitment to angelicin but eliminated recruitment to psoralen. Our results demonstrate an efficient ICL repair pathway in G(1) phase cells dependent on XPC, with entry of DDB2 only after repair synthesis that completes the first repair cycle. DDB2 accumulation at sites of cross-link repair is a marker for the start of the second repair cycle.
Collapse
Affiliation(s)
- Parameswary A Muniandy
- Laboratory of Molecular Gerontology, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Dennis Thapa
- Laboratory of Molecular Gerontology, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | | | | | - Michael M Seidman
- Laboratory of Molecular Gerontology, NIA, National Institutes of Health, Baltimore, Maryland 21224.
| |
Collapse
|
191
|
Minko IG, Kozekov ID, Harris TM, Rizzo CJ, Lloyd RS, Stone MP. Chemistry and biology of DNA containing 1,N(2)-deoxyguanosine adducts of the alpha,beta-unsaturated aldehydes acrolein, crotonaldehyde, and 4-hydroxynonenal. Chem Res Toxicol 2009; 22:759-78. [PMID: 19397281 PMCID: PMC2685875 DOI: 10.1021/tx9000489] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The α,β-unsaturated aldehydes (enals) acrolein, crotonaldehyde, and trans-4-hydroxynonenal (4-HNE) are products of endogenous lipid peroxidation, arising as a consequence of oxidative stress. The addition of enals to dG involves Michael addition of the N2-amine to give N2-(3-oxopropyl)-dG adducts, followed by reversible cyclization of N1 with the aldehyde, yielding 1,N2-dG exocyclic products. The 1,N2-dG exocyclic adducts from acrolein, crotonaldehyde, and 4-HNE exist in human and rodent DNA. The enal-induced 1,N2-dG lesions are repaired by the nucleotide excision repair pathway in both Escherichia coli and mammalian cells. Oligodeoxynucleotides containing structurally defined 1,N2-dG adducts of acrolein, crotonaldehyde, and 4-HNE were synthesized via a postsynthetic modification strategy. Site-specific mutagenesis of enal adducts has been carried out in E. coli and various mammalian cells. In all cases, the predominant mutations observed are G→T transversions, but these adducts are not strongly miscoding. When placed into duplex DNA opposite dC, the 1,N2-dG exocyclic lesions undergo ring opening to the corresponding N2-(3-oxopropyl)-dG derivatives. Significantly, this places a reactive aldehyde in the minor groove of DNA, and the adducted base possesses a modestly perturbed Watson−Crick face. Replication bypass studies in vitro indicate that DNA synthesis past the ring-opened lesions can be catalyzed by pol η, pol ι, and pol κ. It also can be accomplished by a combination of Rev1 and pol ζ acting sequentially. However, efficient nucleotide insertion opposite the 1,N2-dG ring-closed adducts can be carried out only by pol ι and Rev1, two DNA polymerases that do not rely on the Watson−Crick pairing to recognize the template base. The N2-(3-oxopropyl)-dG adducts can undergo further chemistry, forming interstrand DNA cross-links in the 5′-CpG-3′ sequence, intrastrand DNA cross-links, or DNA−protein conjugates. NMR and mass spectrometric analyses indicate that the DNA interstand cross-links contain a mixture of carbinolamine and Schiff base, with the carbinolamine forms of the linkages predominating in duplex DNA. The reduced derivatives of the enal-mediated N2-dG:N2-dG interstrand cross-links can be processed in mammalian cells by a mechanism not requiring homologous recombination. Mutations are rarely generated during processing of these cross-links. In contrast, the reduced acrolein-mediated N2-dG peptide conjugates can be more mutagenic than the corresponding monoadduct. DNA polymerases of the DinB family, pol IV in E. coli and pol κ in human, are implicated in error-free bypass of model acrolein-mediated N2-dG secondary adducts, the interstrand cross-links, and the peptide conjugates.
Collapse
Affiliation(s)
- Irina G Minko
- Center for Research on Occupational and Environmental Toxicology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | | | | | | | | | |
Collapse
|
192
|
Prognostic and Predictive Markers of Benefit from Adjuvant Chemotherapy in Early-Stage Non-small Cell Lung Cancer. J Thorac Oncol 2009; 4:891-910. [DOI: 10.1097/jto.0b013e3181a4b8fb] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
193
|
Steffensen KD, Waldstrøm M, Jakobsen A. The relationship of platinum resistance and ERCC1 protein expression in epithelial ovarian cancer. Int J Gynecol Cancer 2009; 19:820-5. [PMID: 19574766 DOI: 10.1111/igc.0b013e3181a12e09] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Although platinum-based chemotherapy remains the cornerstone for treatment of ovarian cancer, some patients are resistant to the treatment and will therefore not benefit from the standard platinum-based chemotherapy. Preclinical and clinical data have suggested a potential use of excision repair cross-complementation group 1 enzyme (ERCC1) as a molecular predictor of clinical resistance to platinum-based chemotherapy. Excision repair cross-complementation group 1 enzyme is a key enzyme in the nucleotide excision repair pathway which is involved in the DNA repair mechanisms in tumor cells damaged by treatment with platinum agents. The primary aim of the present study was to investigate if immunohistochemical expression of ERCC1 protein was associated with resistance to standard combination carboplatin and paclitaxel chemotherapy in newly diagnosed ovarian cancer patients. METHODS Formalin-fixed, paraffin-embedded tissue sections from 101 patients with newly diagnosed ovarian cancer were used for immunohistochemical staining for the ERCC1 protein. All patients received carboplatin-paclitaxel combination chemotherapy. RESULTS Excision repair cross-complementation group 1 enzyme protein overexpression was found in 13.9% of the tumors. Platinum resistance was found in 75% of the tumors with positive ERCC1 protein expression compared with 27% among the patients with negative tumor staining for ERCC1 (P = 0.0013). These findings translated into a significant difference in progression-free survival in both univariate (P = 0.0012) and in multivariate analysis (P = 0.006). CONCLUSIONS The data presented suggest a positive association between positive ERCC1 protein expression and clinical resistance to platinum-based chemotherapy.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/drug therapy
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/pathology
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carboplatin/administration & dosage
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- DNA-Binding Proteins/metabolism
- Drug Resistance, Neoplasm
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/pathology
- Endonucleases/metabolism
- Female
- Humans
- Immunoenzyme Techniques
- Middle Aged
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Paclitaxel/administration & dosage
- Prognosis
- Survival Rate
- Treatment Outcome
Collapse
|
194
|
Gu Y, Patterson AV, Atwell GJ, Chernikova SB, Brown JM, Thompson LH, Wilson WR. Roles of DNA repair and reductase activity in the cytotoxicity of the hypoxia-activated dinitrobenzamide mustard PR-104A. Mol Cancer Ther 2009; 8:1714-23. [PMID: 19509245 DOI: 10.1158/1535-7163.mct-08-1209] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PR-104 is a dinitrobenzamide mustard currently in clinical trial as a hypoxia-activated prodrug. Its major metabolite, PR-104A, is metabolized to the corresponding hydroxylamine (PR-104H) and amine (PR-104M), resulting in activation of the nitrogen mustard moiety. We characterize DNA damage responsible for cytotoxicity of PR-104A by comparing sensitivity of repair-defective hamster Chinese hamster ovary cell lines with their repair-competent counterparts. PR-104H showed a repair profile similar to the reference DNA cross-linking agents chlorambucil and mitomycin C, with marked hypersensitivity of XPF(-/-), ERCC1(-/-), and Rad51D(-/-) cells but not of XPD(-/-) or DNA-PK(CS)(-/-) cells. This pattern confirmed the expected dependence on the ERCC1-XPF endonuclease, implicated in unhooking DNA interstrand cross-links at blocked replication forks, and homologous recombination repair (HRR) in restarting collapsed forks. However, even under anoxia, the hypersensitivity of XPF(-/-), ERCC1(-/-), and Rad51D(-/-) cells to PR-104A itself was lower than for chlorambucil. To test whether this reflects inefficient PR-104A reduction, a soluble form of human NADPH:cytochrome P450 oxidoreductase was stably expressed in Rad51D(-/-) cells and their HRR-restored counterpart. This expression increased hypoxic metabolism of PR-104A to PR-104H and PR-104M as well as hypoxia-selective cytotoxicity of PR-104A and its dependence on HRR. We conclude that PR-104A cytotoxicity is primarily due to DNA interstrand cross-linking by its reduced metabolites, although under conditions of inefficient PR-104A reduction (low reductase expression or aerobic cells), a second mechanism contributes to cell killing. This study shows that hypoxia, reductase activity, and DNA interstrand cross-link repair proficiency are key variables that interact to determine PR-104A sensitivity.
Collapse
Affiliation(s)
- Yongchuan Gu
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
195
|
Zhang HB, Xue JJ, Zhao XL, Liu DG, Li Y. Synthesis and biological evaluation of novel steroid-linked nitrogen mustards. CHINESE CHEM LETT 2009. [DOI: 10.1016/j.cclet.2009.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
196
|
Lomonaco SL, Xu XS, Wang G. The role of Bcl-x(L) protein in nucleotide excision repair-facilitated cell protection against cisplatin-induced apoptosis. DNA Cell Biol 2009; 28:285-94. [PMID: 19317621 PMCID: PMC2903458 DOI: 10.1089/dna.2008.0815] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 12/21/2008] [Accepted: 12/22/2008] [Indexed: 11/13/2022] Open
Abstract
Many anticancer drugs target the genomic DNA of cancer cells by generating DNA damage and inducing apoptosis. DNA repair protects cells against DNA damage-induced apoptosis. Although the mechanisms of DNA repair and apoptosis have been extensively studied, the mechanism by which DNA repair prevents DNA damage-induced apoptosis is not fully understood. We studied the role of the antiapoptotic Bcl-x(L) protein in nucleotide excision repair (NER)-facilitated cell protection against cisplatin-induced apoptosis. Using both normal human fibroblasts (NF) and NER-defective xeroderma pigmentosum group A (XPA) and group G (XPG) fibroblasts, we demonstrated that a functional NER is required for cisplatin-induced transcription of the bcl-x(l) gene. The results obtained from our Western blots revealed that the cisplatin treatment led to an increase in the level of Bcl-x(L) protein in NF cells, but a decrease in the level of Bcl-x(L) protein in both XPA and XPG cells. The results of our immunofluorescence staining indicated that a functional NER pathway was required for cisplatin-induced translocation of NF-kappaB p65 from cytoplasm into nucleus, indicative of NF-kappaB activation. Given the important function of NF-kappaB in regulating transcription of the bcl-x(l) gene and the Bcl-x(L) protein in preventing apoptosis, these results suggest that NER may protect cells against cisplatin-induced apoptosis by activating NF-kappaB, which further induces transcription of the bcl-x(l) gene, resulting in an accumulation of Bcl-x(L) protein and activation of the cell survival pathway that leads to increased cell survival under cisplatin treatment.
Collapse
Affiliation(s)
- Stephanie L Lomonaco
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
197
|
Zhao J, Jain A, Iyer RR, Modrich PL, Vasquez KM. Mismatch repair and nucleotide excision repair proteins cooperate in the recognition of DNA interstrand crosslinks. Nucleic Acids Res 2009; 37:4420-9. [PMID: 19468048 PMCID: PMC2715249 DOI: 10.1093/nar/gkp399] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
DNA interstrand crosslinks (ICLs) are among the most cytotoxic types of DNA damage, thus ICL-inducing agents such as psoralen, are clinically useful chemotherapeutics. Psoralen-modified triplex-forming oligonucleotides (TFOs) have been used to target ICLs to specific genomic sites to increase the selectivity of these agents. However, how TFO-directed psoralen ICLs (Tdp-ICLs) are recognized and processed in human cells is unclear. Previously, we reported that two essential nucleotide excision repair (NER) protein complexes, XPA–RPA and XPC–RAD23B, recognized ICLs in vitro, and that cells deficient in the DNA mismatch repair (MMR) complex MutSβ were sensitive to psoralen ICLs. To further investigate the role of MutSβ in ICL repair and the potential interaction between proteins from the MMR and NER pathways on these lesions, we performed electrophoretic mobility-shift assays and chromatin immunoprecipitation analysis of MutSβ and NER proteins with Tdp-ICLs. We found that MutSβ bound to Tdp-ICLs with high affinity and specificity in vitro and in vivo, and that MutSβ interacted with XPA–RPA or XPC–RAD23B in recognizing Tdp-ICLs. These data suggest that proteins from the MMR and NER pathways interact in the recognition of ICLs, and provide a mechanistic link by which proteins from multiple repair pathways contribute to ICL repair.
Collapse
Affiliation(s)
- Junhua Zhao
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science-Park Research Division, Smithville, TX 78957, USA
| | | | | | | | | |
Collapse
|
198
|
Episkopou H, Kyrtopoulos SA, Sfikakis PP, Fousteri M, Dimopoulos MA, Mullenders LH, Souliotis VL. Association between Transcriptional Activity, Local Chromatin Structure, and the Efficiencies of Both Subpathways of Nucleotide Excision Repair of Melphalan Adducts. Cancer Res 2009; 69:4424-33. [DOI: 10.1158/0008-5472.can-08-3489] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
199
|
Couvé S, Macé-Aimé G, Rosselli F, Saparbaev MK. The human oxidative DNA glycosylase NEIL1 excises psoralen-induced interstrand DNA cross-links in a three-stranded DNA structure. J Biol Chem 2009; 284:11963-70. [PMID: 19258314 PMCID: PMC2673265 DOI: 10.1074/jbc.m900746200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Previously, we have demonstrated that human oxidative DNA glycosylase NEIL1 excises photoactivated psoralen-induced monoadducts but not genuine interstrand cross-links (ICLs) in duplex DNA. It has been postulated that the repair of ICLs in mammalian cells is mainly linked to DNA replication and proceeds via dual incisions in one DNA strand that bracket the cross-linked site. This process, known as "unhooking," enables strand separation and translesion DNA synthesis through the gap, yielding a three-stranded DNA repair intermediate composed of a short unhooked oligomer covalently bound to the duplex. At present, the detailed molecular mechanism of ICL repair in mammalian cells remains unclear. Here, we constructed and characterized three-stranded DNA structures containing a single ICL as substrates for the base excision repair proteins. We show that NEIL1 excises with high efficiency the unhooked ICL fragment within a three-stranded DNA structure. Complete reconstitution of the repair of unhooked ICL shows that it can be processed in a short patch base excision repair pathway. The new substrate specificity of NEIL1 points to a preferential involvement in the replication-associated repair of ICLs. Based on these data, we propose a model for the mechanism of ICL repair in mammalian cells that implicates the DNA glycosylase activity of NEIL1 downstream of Xeroderma Pigmentosum group F/Excision Repair Cross-Complementing 1 endonuclease complex (XPF/ERCC1) and translesion DNA synthesis repair steps. Finally, our data demonstrate that Nei-like proteins from Escherichia coli to human cells can excise bulky unhooked psoralen-induced ICLs via hydrolysis of glycosidic bond between cross-linked base and deoxyribose sugar, thus providing an alternative heuristic solution for the removal of complex DNA lesions.
Collapse
Affiliation(s)
- Sophie Couvé
- Groupe "Réparation de l'ADN," CNRS UMR 8126, F-94805 Villejuif Cedex, France
| | | | | | | |
Collapse
|
200
|
Landgraeber S, von Knoch M, Löer F, Wegner A, Tsokos M, Schmid KW, Totsch M. ERCC1 expression in aseptic loosening after total hip replacement. J Biomed Mater Res A 2009; 92:556-62. [PMID: 19235214 DOI: 10.1002/jbm.a.32391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Particle-induced osteolysis is a major cause of aseptic loosening after total joint replacement. The purpose of the current study was to evaluate the DNA damage repair capacity of macrophages in patients with aseptic hip loosening by determination of ERCC1. Moreover, we wanted to elucidate if the potency of the DNA-repair mechanisms correlates with the survival of joint implants. For this purpose we compared the immunohistochemical ERCC1 expression in capsules and interface membranes of patients with loosening of a hip replacement in the first 10 years after implantation with those in patients with late loosening. In analogy with ERCC1 studies on cancer in humans we calculated the semi-quantitative H-score by multiplying the staining intensity with the proportion score of positive stained macrophages. The level of ERCC1 reaction in the specimens taken from patients with early aseptic loosening (mean H-score 0.57) was clearly lower in comparison with those from patients undergoing exchange hip arthroplasty later than 10 years after surgery (mean H-score 2.24). We determined an H-score for ERCC1 expression of 1 as a cutoff point giving a sensitivity and specificity of 100% for identification of early aseptic loosening after less than 10 years. In summary, lower levels of ERCC1 were found in patients with early aseptic loosening compared to patients with aseptic loosening later than 10 years.
Collapse
Affiliation(s)
- Stefan Landgraeber
- Department of Orthopaedics, University of Duisburg-Essen, Pattbergstrasse 1-3, 45239 Essen, Germany.
| | | | | | | | | | | | | |
Collapse
|