1
|
Gutierrez R, Chan AYS, Lai SWT, Itoh S, Lee DH, Sun K, Battad A, Chen S, O'Connor TR, Shuck SC. Lack of mismatch repair enhances resistance to methylating agents for cells deficient in oxidative demethylation. J Biol Chem 2024; 300:107492. [PMID: 38925328 PMCID: PMC11326903 DOI: 10.1016/j.jbc.2024.107492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The human alkylation B (AlkB) homologs, ALKBH2 and ALKBH3, respond to methylation damage to maintain genomic integrity and cellular viability. Both ALKBH2 and ALKBH3 are direct reversal repair enzymes that remove 1-methyladenine (1meA) and 3-methylcytosine (3meC) lesions commonly generated by alkylating chemotherapeutic agents. Thus, the existence of deficiencies in ALKBH proteins can be exploited in synergy with chemotherapy. In this study, we investigated possible interactions between ALKBH2 and ALKBH3 with other proteins that could alter damage response and discovered an interaction with the mismatch repair (MMR) system. To test whether the lack of active MMR impacts ALKBH2 and/or ALKBH3 response to methylating agents, we generated cells deficient in ALKBH2, ALKBH3, or both in addition to Mlh homolog 1 (MLH1), another MMR protein. We found that MLH1koALKBH3ko cells showed enhanced resistance toward SN1- and SN2-type methylating agents, whereas MLH1koALKBH2ko cells were only resistant to SN1-type methylating agents. Concomitant loss of ALKBH2 and ALKBH3 (ALKBH2ko3ko) rendered cells sensitive to SN1- and SN2-agents, but the additional loss of MLH1 enhanced resistance to both types of damage. We also showed that ALKBH2ko3ko cells have an ATR-dependent arrest at the G2/M checkpoint, increased apoptotic signaling, and replication fork stress in response to methylation. However, these responses were not observed with the loss of functional MLH1 in MLH1koALKBH2ko3ko cells. Finally, in MLH1koALKBH2ko3ko cells, we observed elevated mutant frequency in untreated and temozolomide treated cells. These results suggest that obtaining a more accurate prognosis of chemotherapeutic outcome requires information on the functionality of ALKBH2, ALKBH3, and MLH1.
Collapse
Affiliation(s)
- Roberto Gutierrez
- Department of Cancer Biology, Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Annie Yin S Chan
- Department of Cancer Biology, Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Seigmund Wai Tsuen Lai
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Shunsuke Itoh
- Department of Cancer Biology, Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Dong-Hyun Lee
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, South Korea
| | - Kelani Sun
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Alana Battad
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Shiuan Chen
- Department of Cancer Biology, Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute at the City of Hope, Duarte, California, USA
| | - Timothy R O'Connor
- Department of Cancer Biology, Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute at the City of Hope, Duarte, California, USA.
| | - Sarah C Shuck
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute at the City of Hope, Duarte, California, USA.
| |
Collapse
|
2
|
Kumar A, Mathew V, Stirling PC. Dynamics of DNA damage-induced nuclear inclusions are regulated by SUMOylation of Btn2. Nat Commun 2024; 15:3215. [PMID: 38615096 PMCID: PMC11016081 DOI: 10.1038/s41467-024-47615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 04/05/2024] [Indexed: 04/15/2024] Open
Abstract
Spatial compartmentalization is a key facet of protein quality control that serves to store disassembled or non-native proteins until triage to the refolding or degradation machinery can occur in a regulated manner. Yeast cells sequester nuclear proteins at intranuclear quality control bodies (INQ) in response to various stresses, although the regulation of this process remains poorly understood. Here we reveal the SUMO modification of the small heat shock protein Btn2 under DNA damage and place Btn2 SUMOylation in a pathway promoting protein clearance from INQ structures. Along with other chaperones, and degradation machinery, Btn2-SUMO promotes INQ clearance from cells recovering from genotoxic stress. These data link small heat shock protein post-translational modification to the regulation of protein sequestration in the yeast nucleus.
Collapse
Affiliation(s)
- Arun Kumar
- Terry Fox Laboratory, BC Cancer, 675 West 10th Avenue, Vancouver, BC, V5Z1L3, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| | - Veena Mathew
- Terry Fox Laboratory, BC Cancer, 675 West 10th Avenue, Vancouver, BC, V5Z1L3, Canada
| | - Peter C Stirling
- Terry Fox Laboratory, BC Cancer, 675 West 10th Avenue, Vancouver, BC, V5Z1L3, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T1Z4, Canada.
| |
Collapse
|
3
|
Her J, Zheng H, Bunting SF. RNF4 sustains Myc-driven tumorigenesis by facilitating DNA replication. J Clin Invest 2024; 134:e167419. [PMID: 38530355 PMCID: PMC11093604 DOI: 10.1172/jci167419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
The mammalian SUMO-targeted E3 ubiquitin ligase Rnf4 has been reported to act as a regulator of DNA repair, but the importance of RNF4 as a tumor suppressor has not been tested. Using a conditional-knockout mouse model, we deleted Rnf4 in the B cell lineage to test the importance of RNF4 for growth of somatic cells. Although Rnf4-conditional-knockout B cells exhibited substantial genomic instability, Rnf4 deletion caused no increase in tumor susceptibility. In contrast, Rnf4 deletion extended the healthy lifespan of mice expressing an oncogenic c-myc transgene. Rnf4 activity is essential for normal DNA replication, and in its absence, there was a failure in ATR-CHK1 signaling of replication stress. Factors that normally mediate replication fork stability, including members of the Fanconi anemia gene family and the helicases PIF1 and RECQL5, showed reduced accumulation at replication forks in the absence of RNF4. RNF4 deficiency also resulted in an accumulation of hyper-SUMOylated proteins in chromatin, including members of the SMC5/6 complex, which contributes to replication failure by a mechanism dependent on RAD51. These findings indicate that RNF4, which shows increased expression in multiple human tumor types, is a potential target for anticancer therapy, especially in tumors expressing c-myc.
Collapse
Affiliation(s)
- Joonyoung Her
- Department of Molecular Biology and Biochemistry and
| | - Haiyan Zheng
- Biological Mass Spectrometry Facility, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | | |
Collapse
|
4
|
Gaspary A, Laureau R, Dyatel A, Dursuk G, Simon Y, Berchowitz LE. Rie1 and Sgn1 form an RNA-binding complex that enforces the meiotic entry cell fate decision. J Cell Biol 2023; 222:e202302074. [PMID: 37638885 PMCID: PMC10460998 DOI: 10.1083/jcb.202302074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/28/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
Budding yeast cells have the capacity to adopt few but distinct physiological states depending on environmental conditions. Vegetative cells proliferate rapidly by budding while spores can survive prolonged periods of nutrient deprivation and/or desiccation. Whether or not a yeast cell will enter meiosis and sporulate represents a critical decision that could be lethal if made in error. Most cell fate decisions, including those of yeast, are understood as being triggered by the activation of master transcription factors. However, mechanisms that enforce cell fates posttranscriptionally have been more difficult to attain. Here, we perform a forward genetic screen to determine RNA-binding proteins that affect meiotic entry at the posttranscriptional level. Our screen revealed several candidates with meiotic entry phenotypes, the most significant being RIE1, which encodes an RRM-containing protein. We demonstrate that Rie1 binds RNA, is associated with the translational machinery, and acts posttranscriptionally to enhance protein levels of the master transcription factor Ime1 in sporulation conditions. We also identified a physical binding partner of Rie1, Sgn1, which is another RRM-containing protein that plays a role in timely Ime1 expression. We demonstrate that these proteins act independently of cell size regulation pathways to promote meiotic entry. We propose a model explaining how constitutively expressed RNA-binding proteins, such as Rie1 and Sgn1, can act in cell fate decisions both as switch-like enforcers and as repressors of spurious cell fate activation.
Collapse
Affiliation(s)
- Alec Gaspary
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Raphaelle Laureau
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Annie Dyatel
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Gizem Dursuk
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Yael Simon
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Luke E. Berchowitz
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer’s and the Aging Brain, New York, NY, USA
| |
Collapse
|
5
|
Bercu JP, Zhang S, Sobol Z, Escobar PA, Van P, Schuler M. Comparison of the transgenic rodent mutation assay, error corrected next generation duplex sequencing, and the alkaline comet assay to detect dose-related mutations following exposure to N-nitrosodiethylamine. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2023; 891:503685. [PMID: 37770142 DOI: 10.1016/j.mrgentox.2023.503685] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023]
Abstract
N-Nitrosodiethylamine (NDEA), a well-studied N-nitrosamine, was tested in rats to compare the dose-response relationship of three genotoxicity endpoints. Mutant / mutation frequencies were determined using the transgenic rodent (TGR) gene mutation assay and error corrected next generation sequencing (ecNGS) (i.e., duplex sequencing (DS)), and genetic damage was detected by the alkaline comet assay. Big Blue® (cII Locus) animals (n = 6 per dose group) were administered doses of 0.001, 0.01, 0.1, 1, 3 mg/kg/day NDEA by oral gavage. Samples were collected for cII mutation and DS analyses following 28-days of exposure and 3 days recovery. In a separate study, male Sprague-Dawley (SD) rats (n = 6 per dose group) were administered the same doses by oral gavage for two consecutive days and then samples collected for the alkaline comet assay. A dose-related increase in mutant / mutation frequencies of the liver but not duodenum was observed using the TGR assay and DS with DS resulting in a slightly more sensitive response, with a lower benchmark dose (BMD). In addition, a dose-related increase in percent tail DNA was observed in the liver using the alkaline comet assay. Therefore, DS and comet assays showed good utility for hazard identification and dose-response analysis of a representative N-nitrosamine comparable to the TGR gene mutation assay.
Collapse
Affiliation(s)
- Joel P Bercu
- Gilead Sciences, Inc., Nonclinical Safety and Pathobiology (NSP), Foster City, CA 94404, USA.
| | - Shaofei Zhang
- Pfizer Research, Development, and Medical, Groton, CT, USA.
| | | | | | - Phu Van
- TwinStrand Biosciences, Inc., Seattle, WA, USA
| | - Maik Schuler
- Pfizer Research, Development, and Medical, Groton, CT, USA
| |
Collapse
|
6
|
Fahrer J, Christmann M. DNA Alkylation Damage by Nitrosamines and Relevant DNA Repair Pathways. Int J Mol Sci 2023; 24:ijms24054684. [PMID: 36902118 PMCID: PMC10003415 DOI: 10.3390/ijms24054684] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/17/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Nitrosamines occur widespread in food, drinking water, cosmetics, as well as tobacco smoke and can arise endogenously. More recently, nitrosamines have been detected as impurities in various drugs. This is of particular concern as nitrosamines are alkylating agents that are genotoxic and carcinogenic. We first summarize the current knowledge on the different sources and chemical nature of alkylating agents with a focus on relevant nitrosamines. Subsequently, we present the major DNA alkylation adducts induced by nitrosamines upon their metabolic activation by CYP450 monooxygenases. We then describe the DNA repair pathways engaged by the various DNA alkylation adducts, which include base excision repair, direct damage reversal by MGMT and ALKBH, as well as nucleotide excision repair. Their roles in the protection against the genotoxic and carcinogenic effects of nitrosamines are highlighted. Finally, we address DNA translesion synthesis as a DNA damage tolerance mechanism relevant to DNA alkylation adducts.
Collapse
Affiliation(s)
- Jörg Fahrer
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Erwin-Schrödinger Strasse 52, D-67663 Kaiserslautern, Germany
- Correspondence: (J.F.); (M.C.); Tel.: +496312052974 (J.F.); Tel: +496131179066 (M.C.)
| | - Markus Christmann
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Correspondence: (J.F.); (M.C.); Tel.: +496312052974 (J.F.); Tel: +496131179066 (M.C.)
| |
Collapse
|
7
|
Bankoglu EE, Schuele C, Stopper H. Cell survival after DNA damage in the comet assay. Arch Toxicol 2021; 95:3803-3813. [PMID: 34609522 PMCID: PMC8536587 DOI: 10.1007/s00204-021-03164-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/16/2021] [Indexed: 11/26/2022]
Abstract
The comet assay is widely used in basic research, genotoxicity testing, and human biomonitoring. However, interpretation of the comet assay data might benefit from a better understanding of the future fate of a cell with DNA damage. DNA damage is in principle repairable, or if extensive, can lead to cell death. Here, we have correlated the maximally induced DNA damage with three test substances in TK6 cells with the survival of the cells. For this, we selected hydrogen peroxide (H2O2) as an oxidizing agent, methyl methanesulfonate (MMS) as an alkylating agent and etoposide as a topoisomerase II inhibitor. We measured cell viability, cell proliferation, apoptosis, and micronucleus frequency on the following day, in the same cell culture, which had been analyzed in the comet assay. After treatment, a concentration dependent increase in DNA damage and in the percentage of non-vital and apoptotic cells was found for each substance. Values greater than 20-30% DNA in tail caused the death of more than 50% of the cells, with etoposide causing slightly more cell death than H2O2 or MMS. Despite that, cells seemed to repair of at least some DNA damage within few hours after substance removal. Overall, the reduction of DNA damage over time is due to both DNA repair and death of heavily damaged cells. We recommend that in experiments with induction of DNA damage of more than 20% DNA in tail, survival data for the cells are provided.
Collapse
Affiliation(s)
- Ezgi Eyluel Bankoglu
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Straße 9, 97078, Wuerzburg, Germany
| | - Carolin Schuele
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Straße 9, 97078, Wuerzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Straße 9, 97078, Wuerzburg, Germany.
| |
Collapse
|
8
|
Causes and consequences of DNA damage-induced autophagy. Matrix Biol 2021; 100-101:39-53. [DOI: 10.1016/j.matbio.2021.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023]
|
9
|
Bansod S, Bung N, Singh P, Suthram N, Choudhury H, Roy A, Bulusu G, Bhattacharyya S. Elucidation of an essential function of the unique charged domain of Plasmodium topoisomerase III. Biochem J 2020; 477:BCJ20200318. [PMID: 33241842 DOI: 10.1042/bcj20200318] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/27/2020] [Accepted: 11/26/2020] [Indexed: 12/28/2022]
Abstract
Topoisomerase III (TopoIII) along with RecQ helicases are required for the resolution of abnormal DNA structures that result from the stalling of replication forks. Sequence analyses have identified a putative TopoIII in the Plasmodium falciparum genome (PfTopoIII). PfTopoIII shows dual nuclear and mitochondrial localization. The expression and association of PfTopoIII with mtDNA is tightly linked to the asexual replication of the parasite. In this study, we observed that PfTopoIII physically interacts with PfBlm and PfWrn. Sequence alignment and domain analyses have revealed that it contains a unique positively charged region, spanning 85 amino acids, within domain II. A molecular dynamics simulation study revealed that this unstructured domain communicates with DNA and attains a thermodynamically stable state upon DNA binding. Here, we found that the association between PfTopoIII and the mitochondrial genome is negatively affected by the absence of the charged domain. Our study shows that PfTOPOIII can completely rescue the slow growth phenotype of the ΔtopoIII strain in Saccharomyces cerevisiae, but neither PfY421FtopoIII (catalytic-active site mutant) nor Pf(Δ259-337)topoIII (charged region deletion mutant) can functionally complement ScTOPOIII. Hydroxyurea (HU) led to stalling of the replication fork during the S phase, caused moderate toxicity to the growth of P. falciparum, and was associated with concomitant transcriptional upregulation of PfTOPOIII. In addition, ectopic expression of PfTOPOIII reversed HU-induced toxicity. Interestingly, the expression of Pf(Δ259-337)topoIII failed to reverse HU-mediated toxicity. Taken together, our results establish the importance of TopoIII during Plasmodium replication and emphasize the essential requirement of the charged domain in PfTopoIII function.
Collapse
Affiliation(s)
- Shephali Bansod
- University of Hyderabad, School of Life Sciences, Hyderabad, India
| | - Navneet Bung
- Tata Consultancy Services Limited, Hyderabad, India, India
| | - Priyanka Singh
- University of Hyderabad, School of Life Sciences, Hyderabad, India
| | | | | | - Arijit Roy
- Tata Consultancy Services Limited, Hyderabad, India
| | | | | |
Collapse
|
10
|
Langston RE, Palazzola D, Bonnell E, Wellinger RJ, Weinert T. Loss of Cdc13 causes genome instability by a deficiency in replication-dependent telomere capping. PLoS Genet 2020; 16:e1008733. [PMID: 32287268 PMCID: PMC7205313 DOI: 10.1371/journal.pgen.1008733] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 05/07/2020] [Accepted: 03/23/2020] [Indexed: 01/02/2023] Open
Abstract
In budding yeast, Cdc13, Stn1, and Ten1 form the telomere-binding heterotrimer CST complex. Here we investigate the role of Cdc13/CST in maintaining genome stability by using a Chr VII disome system that can generate recombinants, chromosome loss, and enigmatic unstable chromosomes. In cells expressing a temperature sensitive CDC13 allele, cdc13F684S, unstable chromosomes frequently arise from problems in or near a telomere. We found that, when Cdc13 is defective, passage through S phase causes Exo1-dependent ssDNA and unstable chromosomes that are then the source for additional chromosome instability events (e.g. recombinants, chromosome truncations, dicentrics, and/or chromosome loss). We observed that genome instability arises from a defect in Cdc13’s function during DNA replication, not Cdc13’s putative post-replication telomere capping function. The molecular nature of the initial unstable chromosomes formed by a Cdc13-defect involves ssDNA and does not involve homologous recombination nor non-homologous end joining; we speculate the original unstable chromosome may be a one-ended double strand break. This system defines a link between Cdc13’s function during DNA replication and genome stability in the form of unstable chromosomes, that then progress to form other chromosome changes. Eukaryotic chromosomes are linear molecules with specialized end structures called telomeres. Telomeres contain both unique repetitive DNA sequences and specialized proteins that solve several biological problems by differentiating chromosomal ends from internal breaks, thus preventing chromosome instability. When telomeres are defective, the entire chromosome can become unstable and change, causing mutations and pathology (cancer, aging, etc.). Here we study how a defect in specific telomere proteins causes chromosomal rearrangements, using the model organism Saccharomyces cerevisiae (budding or brewer’s yeast). We find that when specific telomere proteins are defective, errors in DNA replication generate a type of damage that likely involves extensive single-stranded DNA that forms inherently unstable chromosomes, subject to many subsequent instances of instability (e.g. allelic recombinants, chromosome loss, truncations, dicentrics). The telomere protein Cdc13 is part of a protein complex called CST that is conserved in most organisms including mammalian cells. The technical capacity of studies in budding yeast allow a detailed, real-time examination of how telomere defects compromise chromosome stability in a single cell cycle, generating lessons likely relevant to how human telomeres keep human chromosomes stable.
Collapse
Affiliation(s)
- Rachel E. Langston
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Dominic Palazzola
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Erin Bonnell
- Department of Microbiology and Infectiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Raymund J. Wellinger
- Department of Microbiology and Infectiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Ted Weinert
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
11
|
Zhan D, Zhang X, Li J, Ding X, Cui Y, Jia J. MTH1 Inhibitor TH287 Suppresses Gastric Cancer Development Through the Regulation of PI3K/AKT Signaling. Cancer Biother Radiopharm 2020; 35:223-232. [PMID: 32077746 DOI: 10.1089/cbr.2019.3031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background: Cancer cells evade oxidative stress through the MutT homologue-1 (MTH1), a member of the Nudix family. MTH1 maintains genome integrity and the viability of tumor cells. A new class of MTH1 inhibitors have attracted interest as anticancer agents, but their mechanisms of action remain poorly characterized. In this study, the authors evaluated the anticancer effects of the MTH1 inhibitor TH287 on gastric cancer (GCa) cells. Materials and Methods: BGC-823 and SGC-7901 cells were treated with TH287 and CCK-8, and colony-forming assays were performed. Cell migration was assessed through Transwell and scratch assays. Apoptotic status was measured via flow cytometry and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolyl-carbocyanine iodide (JC-1) staining. Cell cycle status was assessed by propidium iodide (PI) staining. The expression of PI3K/AKT signaling-related proteins was verified by western blotting. Results: TH287 inhibited cell viability, reduced cell proliferation, inhibited apoptosis, induced G2/M arrest, and suppressed cell migration. A loss of mitochondrial membrane potential and reduced Bcl-2/Bax expression were also observed in TH287-treated cells. These effects were mediated through the inhibition of pro-oncogenic PI3K/AKT signaling. Conclusions: These findings indicate that the MTH1 inhibitor TH287 mediates an array of anticancer effects in GCa cells through its effects on mitochondrial function and PI3K/AKT signaling. Collectively, these data highlight the promise of TH287 as a novel therapeutic option for GCa cells.
Collapse
Affiliation(s)
- Dankai Zhan
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xinxin Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jiahui Li
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Xiaojiao Ding
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - YiXuan Cui
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jianguang Jia
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
12
|
Mason JM, Chan YL, Weichselbaum RW, Bishop DK. Non-enzymatic roles of human RAD51 at stalled replication forks. Nat Commun 2019; 10:4410. [PMID: 31562309 PMCID: PMC6764946 DOI: 10.1038/s41467-019-12297-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/22/2019] [Indexed: 01/24/2023] Open
Abstract
The central recombination enzyme RAD51 has been implicated in replication fork processing and restart in response to replication stress. Here, we use a separation-of-function allele of RAD51 that retains DNA binding, but not D-loop activity, to reveal mechanistic aspects of RAD51’s roles in the response to replication stress. Here, we find that cells lacking RAD51’s enzymatic activity protect replication forks from MRE11-dependent degradation, as expected from previous studies. Unexpectedly, we find that RAD51’s strand exchange activity is not required to convert stalled forks to a form that can be degraded by DNA2. Such conversion was shown previously to require replication fork regression, supporting a model in which fork regression depends on a non-enzymatic function of RAD51. We also show RAD51 promotes replication restart by both strand exchange-dependent and strand exchange-independent mechanisms. RAD51 has been implicated in replication fork processing and restart in response to replication stress. Here, authors reveal mechanistic aspects of non-enzymatic roles of RAD51 for fork reversal and cooperation with FBH1.
Collapse
Affiliation(s)
- Jennifer M Mason
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA. .,Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA. .,Center for Human Genetics, Clemson University, Clemson, SC, USA.
| | - Yuen-Ling Chan
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Ralph W Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Douglas K Bishop
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA. .,Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Du H, Wang P, Li L, Wang Y. Repair and translesion synthesis of O6-alkylguanine DNA lesions in human cells. J Biol Chem 2019; 294:11144-11153. [PMID: 31167778 PMCID: PMC6643039 DOI: 10.1074/jbc.ra119.009054] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
O6-alkyl-2'-deoxyguanosine (O6-alkyl-dG) lesions are among the most mutagenic and prevalent alkylated DNA lesions that are associated with cancer initiation and progression. In this study, using a shuttle vector-based strand-specific PCR-competitive replication and adduct bypass assay in conjunction with tandem MS for product identification, we systematically assessed the repair and replicative bypass of a series of O6-alkyl-dG lesions, with the alkyl group being a Me, Et, nPr, iPr, nBu, iBu, or sBu, in several human cell lines. We found that the extent of replication-blocking effects of these lesions is influenced by the size of the alkyl groups situated on the O6 position of the guanine base. We also noted involvement of distinct DNA repair pathways and translesion synthesis polymerases (Pols) in ameliorating the replication blockage effects elicited by the straight- and branched-chain O6-alkyl-dG lesions. We observed that O6-methylguanine DNA methyltransferase is effective in removing the smaller alkyl groups from the O6 position of guanine, whereas repair of the branched-chain lesions relied on nucleotide excision repair. Moreover, these lesions were highly mutagenic during cellular replication and exclusively directed G→A mutations; Pol η and Pol ζ participated in error-prone bypass of the straight-chain lesions, whereas Pol κ preferentially incorporated the correct dCMP opposite the branched-chain lesions. Together, these results uncover key cellular proteins involved in repair and translesion synthesis of O6-alkyl-dG lesions and provide a better understanding of the roles of these types of lesions in the etiology of human cancer.
Collapse
Affiliation(s)
- Hua Du
- Department of Chemistry, University of California, Riverside, California 92521-0403
| | - Pengcheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403
| | - Lin Li
- Department of Chemistry, University of California, Riverside, California 92521-0403
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403
| |
Collapse
|
14
|
Jemth AS, Gustafsson R, Bräutigam L, Henriksson L, Vallin KSA, Sarno A, Almlöf I, Homan E, Rasti A, Warpman Berglund U, Stenmark P, Helleday T. MutT homologue 1 (MTH1) catalyzes the hydrolysis of mutagenic O6-methyl-dGTP. Nucleic Acids Res 2019; 46:10888-10904. [PMID: 30304478 PMCID: PMC6237811 DOI: 10.1093/nar/gky896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022] Open
Abstract
Nucleotides in the free pool are more susceptible to nonenzymatic methylation than those protected in the DNA double helix. Methylated nucleotides like O6-methyl-dGTP can be mutagenic and toxic if incorporated into DNA. Removal of methylated nucleotides from the nucleotide pool may therefore be important to maintain genome integrity. We show that MutT homologue 1 (MTH1) efficiently catalyzes the hydrolysis of O6-methyl-dGTP with a catalytic efficiency similar to that for 8-oxo-dGTP. O6-methyl-dGTP activity is exclusive to MTH1 among human NUDIX proteins and conserved through evolution but not found in bacterial MutT. We present a high resolution crystal structure of human and zebrafish MTH1 in complex with O6-methyl-dGMP. By microinjecting fertilized zebrafish eggs with O6-methyl-dGTP and inhibiting MTH1 we demonstrate that survival is dependent on active MTH1 in vivo. O6-methyl-dG levels are higher in DNA extracted from zebrafish embryos microinjected with O6-methyl-dGTP and inhibition of O6-methylguanine-DNA methyl transferase (MGMT) increases the toxicity of O6-methyl-dGTP demonstrating that O6-methyl-dGTP is incorporated into DNA. MTH1 deficiency sensitizes human cells to the alkylating agent Temozolomide, a sensitization that is more pronounced upon MGMT inhibition. These results expand the cellular MTH1 function and suggests MTH1 also is important for removal of methylated nucleotides from the nucleotide pool.
Collapse
Affiliation(s)
- Ann-Sofie Jemth
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Robert Gustafsson
- Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden
| | - Lars Bräutigam
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Linda Henriksson
- Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden
| | - Karl S A Vallin
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Antonio Sarno
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pathology, St. Olavs Hospital, Trondheim, Norway
| | - Ingrid Almlöf
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Evert Homan
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Azita Rasti
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Ulrika Warpman Berglund
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden.,Sheffield Cancer Centre, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| |
Collapse
|
15
|
Trasviña-Arenas CH, David SS, Delaye L, Azuara-Liceaga E, Brieba LG. Evolution of Base Excision Repair in Entamoeba histolytica is shaped by gene loss, gene duplication, and lateral gene transfer. DNA Repair (Amst) 2019; 76:76-88. [DOI: 10.1016/j.dnarep.2019.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 01/14/2019] [Accepted: 02/19/2019] [Indexed: 12/22/2022]
|
16
|
Bokhari B, Sharma S. Stress Marks on the Genome: Use or Lose? Int J Mol Sci 2019; 20:ijms20020364. [PMID: 30654540 PMCID: PMC6358951 DOI: 10.3390/ijms20020364] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/31/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and the resulting damage to DNA are inevitable consequence of endogenous physiological processes further amplified by cellular responses to environmental exposures. If left unrepaired, oxidative DNA lesions can block essential processes such as transcription and replication or can induce mutations. Emerging data also indicate that oxidative base modifications such as 8-oxoG in gene promoters may serve as epigenetic marks, and/or provide a platform for coordination of the initial steps of DNA repair and the assembly of the transcriptional machinery to launch adequate gene expression alterations. Here, we briefly review the current understanding of oxidative lesions in genome stability maintenance and regulation of basal and inducible transcription.
Collapse
Affiliation(s)
- Bayan Bokhari
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, 520 W Street, NW, Washington, DC 20059, USA.
- Department of Biochemistry, Faculty of Applied Medical Science, Umm Al- Qura University, Makkah 21421, Saudi Arabia.
| | - Sudha Sharma
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, 520 W Street, NW, Washington, DC 20059, USA.
- National Human Genome Center, College of Medicine, Howard University, 2041 Georgia Avenue, NW, Washington, DC 20060, USA.
| |
Collapse
|
17
|
Histone tails decrease N7-methyl-2'-deoxyguanosine depurination and yield DNA-protein cross-links in nucleosome core particles and cells. Proc Natl Acad Sci U S A 2018; 115:E11212-E11220. [PMID: 30429328 DOI: 10.1073/pnas.1813338115] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Monofunctional alkylating agents preferentially react at the N7 position of 2'-deoxyguanosine in duplex DNA. Methylated DNA, such as that produced by methyl methanesulfonate (MMS) and temozolomide, exists for days in organisms. The predominant consequence of N7-methyl-2'-deoxyguanosine (MdG) is widely believed to be abasic site (AP) formation via hydrolysis, a process that is slow in free DNA. Examination of MdG reactivity within nucleosome core particles (NCPs) provided two general observations. MdG depurination rate constants are reduced in NCPs compared with when the identical DNA sequence is free in solution. The magnitude of the decrease correlates with proximity to the positively charged histone tails, and experiments in NCPs containing histone variants reveal that positively charged amino acids are responsible for the decreased rate of abasic site formation from MdG. In addition, the lysine-rich histone tails form DNA-protein cross-links (DPCs) with MdG. Cross-link formation is reversible and is ascribed to nucleophilic attack at the C8 position of MdG. DPC and retarded abasic site formation are observed in NCPs randomly damaged by MMS, indicating that these are general processes. Histone-MdG cross-links were also detected by mass spectrometry in chromatin isolated from V79 Chinese hamster lung cells treated with MMS. The formation of DPCs following damage by a monofunctional alkylating agent has not been reported previously. These observations reveal the possibility that such DPCs may contribute to the cytotoxicity of monofunctional alkylating agents, such as MMS, N-methyl-N-nitrosourea, and temozolomide.
Collapse
|
18
|
Gustafsson NMS, Färnegårdh K, Bonagas N, Ninou AH, Groth P, Wiita E, Jönsson M, Hallberg K, Lehto J, Pennisi R, Martinsson J, Norström C, Hollers J, Schultz J, Andersson M, Markova N, Marttila P, Kim B, Norin M, Olin T, Helleday T. Targeting PFKFB3 radiosensitizes cancer cells and suppresses homologous recombination. Nat Commun 2018; 9:3872. [PMID: 30250201 PMCID: PMC6155239 DOI: 10.1038/s41467-018-06287-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
The glycolytic PFKFB3 enzyme is widely overexpressed in cancer cells and an emerging anti-cancer target. Here, we identify PFKFB3 as a critical factor in homologous recombination (HR) repair of DNA double-strand breaks. PFKFB3 rapidly relocates into ionizing radiation (IR)-induced nuclear foci in an MRN-ATM-γH2AX-MDC1-dependent manner and co-localizes with DNA damage and HR repair proteins. PFKFB3 relocalization is critical for recruitment of HR proteins, HR activity, and cell survival upon IR. We develop KAN0438757, a small molecule inhibitor that potently targets PFKFB3. Pharmacological PFKFB3 inhibition impairs recruitment of ribonucleotide reductase M2 and deoxynucleotide incorporation upon DNA repair, and reduces dNTP levels. Importantly, KAN0438757 induces radiosensitization in transformed cells while leaving non-transformed cells unaffected. In summary, we identify a key role for PFKFB3 enzymatic activity in HR repair and present KAN0438757, a selective PFKFB3 inhibitor that could potentially be used as a strategy for the treatment of cancer.
Collapse
Affiliation(s)
- Nina M S Gustafsson
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden.
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden.
| | - Katarina Färnegårdh
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
- Drug Discovery and Development Platform, Science for Life Laboratory, Department of Organic Chemistry, Stockholm University, Box 1030, S-171 21, Solna, Sweden
| | - Nadilly Bonagas
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | - Anna Huguet Ninou
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Petra Groth
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | - Elisee Wiita
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | | | - Kenth Hallberg
- SARomics Biostructures AB, Medicon Village, SE-223 81, Lund, Sweden
- Sprint Bioscience, 141 57, Huddinge, Sweden
| | - Jemina Lehto
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Rosa Pennisi
- Department of Sciences, Roma Tre University, 446 00146 Rome, Italy
| | | | | | - Jessica Hollers
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Johan Schultz
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | | | | | - Petra Marttila
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden
| | - Baek Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Pharmacy, Kyung-Hee University, 02447, Seoul, South Korea
| | - Martin Norin
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Thomas Olin
- Kancera AB, Karolinska Science Park, 171 48, Solna, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, 171 21, Stockholm, Sweden.
- Sheffield Cancer Centre, Department of Oncology and Metabolism, University of Sheffield, S10 2RX, Sheffield, UK.
| |
Collapse
|
19
|
Odabasi IE, Gencturk E, Puza S, Mutlu S, Ulgen KO. A low cost PS based microfluidic platform to investigate cell cycle towards developing a therapeutic strategy for cancer. Biomed Microdevices 2018; 20:57. [DOI: 10.1007/s10544-018-0302-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Abstract
The health of an organism is intimately linked to its ability to repair damaged DNA. Importantly, DNA repair processes are highly dynamic. This highlights the necessity of characterizing DNA repair in live cells. Advanced genome editing and imaging approaches allow us to visualize damaged DNA and its associated factors in real time. Here, we summarize both established and recent methods that are used to induce DNA damage and visualize damaged DNA and its repair in live cells.
Collapse
Affiliation(s)
- Roxanne Oshidari
- Department of Laboratory Medicine and Pathobiology, University of Toronto, MaRS Centre, West Tower, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, University of Toronto, MaRS Centre, West Tower, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada; Canada Research Chairs Program, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
21
|
Fordham SE, Blair HJ, Elstob CJ, Plummer R, Drew Y, Curtin NJ, Heidenreich O, Pal D, Jamieson D, Park C, Pollard J, Fields S, Milne P, Jackson GH, Marr HJ, Menne T, Jones GL, Allan JM. Inhibition of ATR acutely sensitizes acute myeloid leukemia cells to nucleoside analogs that target ribonucleotide reductase. Blood Adv 2018; 2:1157-1169. [PMID: 29789314 PMCID: PMC5965047 DOI: 10.1182/bloodadvances.2017015214] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/09/2018] [Indexed: 12/20/2022] Open
Abstract
The ataxia telangiectasia and Rad3-related (ATR) protein kinase promotes cancer cell survival by signaling stalled replication forks generated by replication stress, a common feature of many cancers including acute myeloid leukemia (AML). Here we show that the antileukemic activity of the chemotherapeutic nucleoside analogs hydroxyurea and gemcitabine was significantly potentiated by ATR inhibition via a mechanism involving ribonucleotide reductase (RNR) abrogation and inhibition of replication fork progression. When administered in combination with gemcitabine, an inhibitor of the M1 RNR subunit, the ATR inhibitor VX-970, eradicated disseminated leukemia in an orthotopic mouse model, eliciting long-term survival and effective cure. These data identify a synergistic interaction between ATR inhibition and RNR loss that will inform the deployment of small molecule inhibitors for the treatment of AML and other hematologic malignancies.
Collapse
Affiliation(s)
- Sarah E Fordham
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen J Blair
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Claire J Elstob
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ruth Plummer
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Yvette Drew
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicola J Curtin
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Olaf Heidenreich
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Deepali Pal
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Jamieson
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Catherine Park
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John Pollard
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - Scott Fields
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - Paul Milne
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| | - Graham H Jackson
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Helen J Marr
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Tobias Menne
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Gail L Jones
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - James M Allan
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
22
|
Gadhikar MA, Zhang J, Shen L, Rao X, Wang J, Zhao M, Kalu NN, Johnson FM, Byers LA, Heymach J, Hittelman WN, Udayakumar D, Pandita RK, Pandita TK, Pickering CR, Redwood AB, Piwnica-Worms H, Schlacher K, Frederick MJ, Myers JN. CDKN2A/p16 Deletion in Head and Neck Cancer Cells Is Associated with CDK2 Activation, Replication Stress, and Vulnerability to CHK1 Inhibition. Cancer Res 2017; 78:781-797. [PMID: 29229598 DOI: 10.1158/0008-5472.can-17-2802] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/30/2017] [Accepted: 11/30/2017] [Indexed: 11/16/2022]
Abstract
Checkpoint kinase inhibitors (CHKi) exhibit striking single-agent activity in certain tumors, but the mechanisms accounting for hypersensitivity are poorly understood. We screened a panel of 49 established human head and neck squamous cell carcinoma (HNSCC) cell lines and report that nearly 20% are hypersensitive to CHKi monotherapy. Hypersensitive cells underwent early S-phase arrest at drug doses sufficient to inhibit greater than 90% of CHK1 activity. Reduced rate of DNA replication fork progression and chromosomal shattering were also observed, suggesting replication stress as a root causative factor in CHKi hypersensitivity. To explore genomic underpinnings of CHKi hypersensitivity, comparative genomic analysis was performed between hypersensitive cells and cells categorized as least sensitive because they showed drug IC50 value greater than the cell panel median and lacked early S-phase arrest. Novel association between CDKN2A/p16 copy number loss, CDK2 activation, replication stress, and hypersensitivity of HNSCC cells to CHKi monotherapy was found. Restoring p16 in cell lines harboring CDKN2A/p16 genomic deletions alleviated CDK2 activation and replication stress, attenuating CHKi hypersensitivity. Taken together, our results suggest a biomarker-driven strategy for selecting HNSCC patients who may benefit the most from CHKi therapy.Significance: These results suggest a biomarker-driven strategy for selecting HNSCC patients who may benefit the most from therapy with CHK inhibitors. Cancer Res; 78(3); 781-97. ©2017 AACR.
Collapse
Affiliation(s)
- Mayur A Gadhikar
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiexin Zhang
- Department of Biostatistics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Li Shen
- Department of Biostatistics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiayu Rao
- Department of Biostatistics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Biostatistics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mei Zhao
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nene N Kalu
- Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Faye M Johnson
- Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren A Byers
- Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Heymach
- Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Walter N Hittelman
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Durga Udayakumar
- Department of Radiation Oncology, Institute for Academic Medicine, Houston Methodist, Houston, Texas
| | - Raj K Pandita
- Department of Radiation Oncology, Institute for Academic Medicine, Houston Methodist, Houston, Texas
| | - Tej K Pandita
- Department of Radiation Oncology, Institute for Academic Medicine, Houston Methodist, Houston, Texas
| | - Curtis R Pickering
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abena B Redwood
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katharina Schlacher
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mitchell J Frederick
- Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, Texas.
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
23
|
Hagenkort A, Paulin CBJ, Desroses M, Sarno A, Wiita E, Mortusewicz O, Koolmeister T, Loseva O, Jemth AS, Almlöf I, Homan E, Lundbäck T, Gustavsson AL, Scobie M, Helleday T. dUTPase inhibition augments replication defects of 5-Fluorouracil. Oncotarget 2017; 8:23713-23726. [PMID: 28423595 PMCID: PMC5410339 DOI: 10.18632/oncotarget.15785] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 02/06/2017] [Indexed: 11/01/2022] Open
Abstract
The antimetabolite 5-Fluorouracil (5-FU) is used in the treatment of various forms of cancer and has a complex mode of action. Despite 6 decades in clinical application the contribution of 5-FdUTP and dUTP [(5-F)dUTP] and 5-FUTP misincorporation into DNA and RNA respectively, for 5-FU-induced toxicity is still under debate.This study investigates DNA replication defects induced by 5-FU treatment and how (5-F)dUTP accumulation contributes to this effect. We reveal that 5-FU treatment leads to extensive problems in DNA replication fork progression, causing accumulation of cells in S-phase, DNA damage and ultimately cell death. Interestingly, these effects can be reinforced by either depletion or inhibition of the deoxyuridine triphosphatase (dUTPase, also known as DUT), highlighting the importance of (5-F)dUTP accumulation for cytotoxicity.With this study, we not only extend the current understanding of the mechanism of action of 5-FU, but also contribute to the characterization of dUTPase inhibitors. We demonstrate that pharmacological inhibition of dUTPase is a promising approach that may improve the efficacy of 5-FU treatment in the clinic.
Collapse
Affiliation(s)
- Anna Hagenkort
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Cynthia B J Paulin
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Matthieu Desroses
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Sarno
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Elisée Wiita
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Oliver Mortusewicz
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tobias Koolmeister
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Olga Loseva
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ann-Sofie Jemth
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Almlöf
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Evert Homan
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Lundbäck
- Chemical Biology Consortium Sweden, Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anna-Lena Gustavsson
- Chemical Biology Consortium Sweden, Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Scobie
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Helleday
- Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Parvathaneni S, Lu X, Chaudhary R, Lal A, Madhusudan S, Sharma S. RECQ1 expression is upregulated in response to DNA damage and in a p53-dependent manner. Oncotarget 2017; 8:75924-75942. [PMID: 29100281 PMCID: PMC5652675 DOI: 10.18632/oncotarget.18237] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/15/2017] [Indexed: 12/11/2022] Open
Abstract
Sensitivity of cancer cells to DNA damaging chemotherapeutics is determined by DNA repair processes. Consequently, cancer cells may upregulate the expression of certain DNA repair genes as a mechanism to promote chemoresistance. Here, we report that RECQ1, a breast cancer susceptibility gene that encodes the most abundant RecQ helicase in humans, is a p53-regulated gene, potentially acting as a defense against DNA damaging agents. We show that RECQ1 mRNA and protein levels are upregulated upon treatment of cancer cells with a variety of DNA damaging agents including the DNA-alkylating agent methylmethanesulfonate (MMS). The MMS-induced upregulation of RECQ1 expression is p53-dependent as it was observed in p53-proficient but not in isogenic p53-deficient cells. The RECQ1 promoter is bound by endogenous p53 and is responsive to p53 in luciferase reporter assays suggesting that RECQ1 is a direct target of p53. Treatment with the chemotherapeutic drugs temozolomide and fotemustine also increased RECQ1 mRNA levels whereas depletion of RECQ1 enhanced cellular sensitivity to these agents. These results identify a previously unrecognized p53-mediated upregulation of RECQ1 expression in response to DNA damage and implicate RECQ1 in the repair of DNA lesions including those induced by alkylating and other chemotherapeutic agents.
Collapse
Affiliation(s)
- Swetha Parvathaneni
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, NW, Washington, DC, 20059, USA
| | - Xing Lu
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, NW, Washington, DC, 20059, USA
| | - Ritu Chaudhary
- Regulatory RNAs and Cancer Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Srinivasan Madhusudan
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, NG51PB, UK
| | - Sudha Sharma
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, NW, Washington, DC, 20059, USA
| |
Collapse
|
25
|
Bellendir SP, Rognstad DJ, Morris LP, Zapotoczny G, Walton WG, Redinbo MR, Ramsden DA, Sekelsky J, Erie DA. Substrate preference of Gen endonucleases highlights the importance of branched structures as DNA damage repair intermediates. Nucleic Acids Res 2017; 45:5333-5348. [PMID: 28369583 PMCID: PMC5435919 DOI: 10.1093/nar/gkx214] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/16/2017] [Accepted: 03/21/2017] [Indexed: 11/20/2022] Open
Abstract
Human GEN1 and yeast Yen1 are endonucleases with the ability to cleave Holliday junctions (HJs), which are proposed intermediates in recombination. In vivo, GEN1 and Yen1 function secondarily to Mus81, which has weak activity on intact HJs. We show that the genetic relationship is reversed in Drosophila, with Gen mutants having more severe defects than mus81 mutants. In vitro, DmGen, like HsGEN1, efficiently cleaves HJs, 5΄ flaps, splayed arms, and replication fork structures. We find that the cleavage rates for 5΄ flaps are significantly higher than those for HJs for both DmGen and HsGEN1, even in vast excess of enzyme over substrate. Kinetic studies suggest that the difference in cleavage rates results from a slow, rate-limiting conformational change prior to HJ cleavage: formation of a productive dimer on the HJ. Despite the stark difference in vivo that Drosophila uses Gen over Mus81 and humans use MUS81 over GEN1, we find the in vitro activities of DmGen and HsGEN1 to be strikingly similar. These findings suggest that simpler branched structures may be more important substrates for Gen orthologs in vivo, and highlight the utility of using the Drosophila model system to further understand these enzymes.
Collapse
Affiliation(s)
| | | | - Lydia P. Morris
- Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
| | | | | | - Matthew R. Redinbo
- Department of Chemistry, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Sciences, Chapel Hill, NC 27599, USA
| | - Dale A. Ramsden
- Curriculum in Genetics and Molecular Biology, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Department of Biochemistry and Biophysics, Chapel Hill, NC 27599, USA
| | - Jeff Sekelsky
- Curriculum in Genetics and Molecular Biology, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Sciences, Chapel Hill, NC 27599, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dorothy A. Erie
- Department of Chemistry, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Sciences, Chapel Hill, NC 27599, USA
| |
Collapse
|
26
|
Increased genome instability is not accompanied by sensitivity to DNA damaging agents in aged yeast cells. DNA Repair (Amst) 2017; 54:1-7. [PMID: 28384494 DOI: 10.1016/j.dnarep.2017.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 11/21/2022]
Abstract
The budding yeast Saccharomyces cerevisiae divides asymmetrically, producing a new daughter cell from the original mother cell. While daughter cells are born with a full lifespan, a mother cell ages with each cell division and can only generate on average 25 daughter cells before dying. Aged yeast cells exhibit genomic instability, which is also a hallmark of human aging. However, it is unclear how this genomic instability contributes to aging. To shed light on this issue, we investigated endogenous DNA damage in S. cerevisiae during replicative aging and tested for age-dependent sensitivity to exogenous DNA damaging agents. Using live-cell imaging in a microfluidic device, we show that aging yeast cells display an increase in spontaneous Rad52 foci, a marker of endogenous DNA damage. Strikingly, this elevated DNA damage is not accompanied by increased sensitivity of aged yeast cells to genotoxic agents nor by global changes in the proteome or transcriptome that would indicate a specific "DNA damage signature". These results indicate that DNA repair proficiency is not compromised in aged yeast cells, suggesting that yeast replicative aging and age-associated genomic instability is likely not a consequence of an inability to repair DNA damage.
Collapse
|
27
|
Dmowski M, Rudzka J, Campbell JL, Jonczyk P, Fijałkowska IJ. Mutations in the Non-Catalytic Subunit Dpb2 of DNA Polymerase Epsilon Affect the Nrm1 Branch of the DNA Replication Checkpoint. PLoS Genet 2017; 13:e1006572. [PMID: 28107343 PMCID: PMC5291541 DOI: 10.1371/journal.pgen.1006572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 02/03/2017] [Accepted: 01/05/2017] [Indexed: 12/14/2022] Open
Abstract
To preserve genome integrity, the S-phase checkpoint senses damaged DNA or nucleotide depletion and when necessary, arrests replication progression and delays cell division. Previous studies, based on two pol2 mutants have suggested the involvement of DNA polymerase epsilon (Pol ε) in sensing DNA replication accuracy in Saccharomyces cerevisiae. Here we have studied the involvement of Pol ε in sensing proper progression of DNA replication, using a mutant in DPB2, the gene coding for a non-catalytic subunit of Pol ε. Under genotoxic conditions, the dpb2-103 cells progress through S phase faster than wild-type cells. Moreover, the Nrm1-dependent branch of the checkpoint, which regulates the expression of many replication checkpoint genes, is impaired in dpb2-103 cells. Finally, deletion of DDC1 in the dpb2-103 mutant is lethal supporting a model of strand-specific activation of the replication checkpoint. This lethality is suppressed by NRM1 deletion. We postulate that improper activation of the Nrm1-branch may explain inefficient replication checkpoint activation in Pol ε mutants. The viability of living organisms depends on the integrity of their genomes. Each cell has to constantly monitor DNA replication and coordinate it with cell division to avoid genomic instability. This is achieved through pathways known as cell cycle checkpoints. Therefore, upon replication perturbation, DNA synthesis slows down and cell division is delayed. For that, a specific signal is induced and propagated through a mechanism that have already been identified but still need investigations. We have isolated a mutated form of Dpb2, the essential subunit of DNA polymerase epsilon (Pol ε) holoenzyme. This mutated form of Pol ε impairs proper activation of the cellular response to replication stress. We show that yeast cells with mutations in the DPB2 gene fail to activate the Nrm1-regulated branch of the checkpoint, which controls numerous genes expressed in response to replication stress. Moreover, our results support the model of parallel activation of replication checkpoint from the leading and lagging DNA strands. This strongly suggests that Pol ε, the leading strand replicase, is involved in replication checkpoint activation from this strand. Our results contribute to the understanding of mechanisms of cellular response to replication stress, which are necessary to preserve genome stability.
Collapse
Affiliation(s)
- Michał Dmowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences Pawińskiego 5a, Warsaw, POLAND
- * E-mail:
| | - Justyna Rudzka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences Pawińskiego 5a, Warsaw, POLAND
| | - Judith L. Campbell
- Braun Laboratories, California Institute of Technology, Pasadena, CA, United States of America
| | - Piotr Jonczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences Pawińskiego 5a, Warsaw, POLAND
| | - Iwona J. Fijałkowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences Pawińskiego 5a, Warsaw, POLAND
| |
Collapse
|
28
|
Lu F, Wu X, Yin F, Chia-Fang Lee C, Yu M, Mihaylov IS, Yu J, Sun H, Zhang H. Regulation of DNA replication and chromosomal polyploidy by the MLL-WDR5-RBBP5 methyltransferases. Biol Open 2016; 5:1449-1460. [PMID: 27744293 PMCID: PMC5087680 DOI: 10.1242/bio.019729] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
DNA replication licensing occurs on chromatin, but how the chromatin template is regulated for replication remains mostly unclear. Here, we have analyzed the requirement of histone methyltransferases for a specific type of replication: the DNA re-replication induced by the downregulation of either Geminin, an inhibitor of replication licensing protein CDT1, or the CRL4CDT2 ubiquitin E3 ligase. We found that siRNA-mediated reduction of essential components of the MLL-WDR5-RBBP5 methyltransferase complexes including WDR5 or RBBP5, which transfer methyl groups to histone H3 at K4 (H3K4), suppressed DNA re-replication and chromosomal polyploidy. Reduction of WDR5/RBBP5 also prevented the activation of H2AX checkpoint caused by re-replication, but not by ultraviolet or X-ray irradiation; and the components of MLL complexes co-localized with the origin recognition complex (ORC) and MCM2-7 replicative helicase complexes at replication origins to control the levels of methylated H3K4. Downregulation of WDR5 or RBBP5 reduced the methylated H3K4 and suppressed the recruitment of MCM2-7 complexes onto replication origins. Our studies indicate that the MLL complexes and H3K4 methylation are required for DNA replication but not for DNA damage repair. Summary: DNA replication or re-replication of DNA induced after loss of Geminin or CLR4CDT2 is regulated by the methylation activities of the MLL-WDR5-RBBP5 methyltransferases on histone H3 at lysine 4 (H3K4).
Collapse
Affiliation(s)
- Fei Lu
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China Basic Science Division, Nevada Cancer Institute, Las Vegas, NV 89135, USA
| | - Xiaojun Wu
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | - Feng Yin
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | | | - Min Yu
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | - Ivailo S Mihaylov
- Basic Science Division, Nevada Cancer Institute, Las Vegas, NV 89135, USA
| | - Jiekai Yu
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, NV 89154, USA
| | - Hong Sun
- Basic Science Division, Nevada Cancer Institute, Las Vegas, NV 89135, USA Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, NV 89154, USA
| | - Hui Zhang
- Basic Science Division, Nevada Cancer Institute, Las Vegas, NV 89135, USA Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, NV 89154, USA
| |
Collapse
|
29
|
Martino J, Bernstein KA. The Shu complex is a conserved regulator of homologous recombination. FEMS Yeast Res 2016; 16:fow073. [PMID: 27589940 DOI: 10.1093/femsyr/fow073] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2016] [Indexed: 02/06/2023] Open
Abstract
Homologous recombination (HR) is an error-free DNA repair mechanism that maintains genome integrity by repairing double-strand breaks (DSBs). Defects in HR lead to genomic instability and are associated with cancer predisposition. A key step in HR is the formation of Rad51 nucleoprotein filaments which are responsible for the homology search and strand invasion steps that define HR. Recently, the budding yeast Shu complex has emerged as an important regulator of Rad51 along with the other Rad51 mediators including Rad52 and the Rad51 paralogs, Rad55-Rad57. The Shu complex is a heterotetramer consisting of two novel Rad51 paralogs, Psy3 and Csm2, along with Shu1 and a SWIM domain-containing protein, Shu2. Studies done primarily in yeast have provided evidence that the Shu complex regulates HR at several types of DNA DSBs (i.e. replication-associated and meiotic DSBs) and that its role in HR is highly conserved across eukaryotic lineages. This review highlights the main findings of these studies and discusses the proposed specific roles of the Shu complex in many aspects of recombination-mediated DNA repair.
Collapse
Affiliation(s)
- Julieta Martino
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Kara A Bernstein
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| |
Collapse
|
30
|
Rozacky J, Nemec AA, Sweasy JB, Kidane D. Gastric cancer associated variant of DNA polymerase beta (Leu22Pro) promotes DNA replication associated double strand breaks. Oncotarget 2016; 6:24474-87. [PMID: 26090616 PMCID: PMC4695199 DOI: 10.18632/oncotarget.4426] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 05/31/2015] [Indexed: 12/14/2022] Open
Abstract
DNA polymerase beta (Pol β) is a key enzymefor the protection against oxidative DNA lesions via itsrole in base excision repair (BER). Approximately 1/3 of tumors studied to date express Pol β variant proteins, and several tumors overexpress Pol β. Pol β possesses DNA polymerase and dRP lyase activities, both of which are known to be important for efficient BER. The dRP lyase activity resides within the 8kDa amino terminal domain of Pol β, is responsible for removal of the 5′ phosphate group (5′-dRP). The DNA polymerase subsequently fills the gaps. Previously, we demonstrated that the human gastric cancer-associated variant of Pol β (Leu22Pro (L22P)) lacks dRP lyase function in vitro. Here, we report that L22P-expressing cells harbor significantly increased replication associated DNA double strand breaks (DSBs) and defective maintenance of the nascent DNA strand (NDS) during replication stress. Moreover, L22P-expressing cells are sensitive to PARP1 inhibitors, which suggests trapped PARP1 binds to the 5′-dRP group and blocks replications forks, resulting in fork collapse and DSBs. Our data suggest that the normal function of the dRP lyase is critical to maintain replication fork integrity and prevent replication fork collapse to DSBs and cellular transformation.
Collapse
Affiliation(s)
- Jenna Rozacky
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX, USA
| | - Antoni A Nemec
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Joann B Sweasy
- Departments of Therapeutic Radiology and Genetics, The Yale Comprehensive Cancer Center, New Haven CT, USA
| | - Dawit Kidane
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX, USA
| |
Collapse
|
31
|
Janke R, Kong J, Braberg H, Cantin G, Yates JR, Krogan NJ, Heyer WD. Nonsense-mediated decay regulates key components of homologous recombination. Nucleic Acids Res 2016; 44:5218-30. [PMID: 27001511 PMCID: PMC4914092 DOI: 10.1093/nar/gkw182] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 12/29/2022] Open
Abstract
Cells frequently experience DNA damage that requires repair by homologous recombination (HR). Proteins involved in HR are carefully coordinated to ensure proper and efficient repair without interfering with normal cellular processes. In Saccharomyces cerevisiae, Rad55 functions in the early steps of HR and is regulated in response to DNA damage through phosphorylation by the Mec1 and Rad53 kinases of the DNA damage response. To further identify regulatory processes that target HR, we performed a high-throughput genetic interaction screen with RAD55 phosphorylation site mutants. Genes involved in the mRNA quality control process, nonsense-mediated decay (NMD), were found to genetically interact with rad55 phospho-site mutants. Further characterization revealed that RAD55 transcript and protein levels are regulated by NMD. Regulation of HR by NMD extends to multiple targets beyond RAD55, including RAD51, RAD54 and RAD57 Finally, we demonstrate that loss of NMD results in an increase in recombination rates and resistance to the DNA damaging agent methyl methanesulfonate, suggesting this pathway negatively regulates HR under normal growth conditions.
Collapse
Affiliation(s)
- Ryan Janke
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA 95616-8665, USA
| | - Jeremy Kong
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA 95616-8665, USA
| | - Hannes Braberg
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158-2517, USA
| | - Greg Cantin
- Department of Cell Biology, SR-11, Scripps Research institute, La Jolla, CA 92307, USA
| | - John R Yates
- Department of Cell Biology, SR-11, Scripps Research institute, La Jolla, CA 92307, USA
| | - Nevan J Krogan
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158-2517, USA California Institute for Quantitative Biosciences, QB3, San Francisco, CA 94158-2517, USA J. David Gladstone Institute, San Francisco, CA, 94158-2517, USA
| | - Wolf-Dietrich Heyer
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA 95616-8665, USA Department of Molecular & Cellular Biology University of California, Davis, CA 95616-8665, USA
| |
Collapse
|
32
|
Devlin R, Marques CA, Paape D, Prorocic M, Zurita-Leal AC, Campbell SJ, Lapsley C, Dickens N, McCulloch R. Mapping replication dynamics in Trypanosoma brucei reveals a link with telomere transcription and antigenic variation. eLife 2016; 5:e12765. [PMID: 27228154 PMCID: PMC4946898 DOI: 10.7554/elife.12765] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 05/26/2016] [Indexed: 01/14/2023] Open
Abstract
Survival of Trypanosoma brucei depends upon switches in its protective Variant Surface Glycoprotein (VSG) coat by antigenic variation. VSG switching occurs by frequent homologous recombination, which is thought to require locus-specific initiation. Here, we show that a RecQ helicase, RECQ2, acts to repair DNA breaks, including in the telomeric site of VSG expression. Despite this, RECQ2 loss does not impair antigenic variation, but causes increased VSG switching by recombination, arguing against models for VSG switch initiation through direct generation of a DNA double strand break (DSB). Indeed, we show DSBs inefficiently direct recombination in the VSG expression site. By mapping genome replication dynamics, we reveal that the transcribed VSG expression site is the only telomeric site that is early replicating - a differential timing only seen in mammal-infective parasites. Specific association between VSG transcription and replication timing reveals a model for antigenic variation based on replication-derived DNA fragility.
Collapse
Affiliation(s)
- Rebecca Devlin
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Catarina A Marques
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Daniel Paape
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Marko Prorocic
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Andrea C Zurita-Leal
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Samantha J Campbell
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Craig Lapsley
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Nicholas Dickens
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Richard McCulloch
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
33
|
Naylor RM, Jeganathan KB, Cao X, van Deursen JM. Nuclear pore protein NUP88 activates anaphase-promoting complex to promote aneuploidy. J Clin Invest 2016; 126:543-59. [PMID: 26731471 DOI: 10.1172/jci82277] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
The nuclear pore complex protein NUP88 is frequently elevated in aggressive human cancers and correlates with reduced patient survival; however, it is unclear whether and how NUP88 overexpression drives tumorigenesis. Here, we show that mice overexpressing NUP88 are cancer prone and form intestinal tumors. To determine whether overexpression of NUP88 drives tumorigenesis, we engineered transgenic mice with doxycycline-inducible expression of Nup88. Surprisingly, NUP88 overexpression did not alter global nuclear transport, but was a potent inducer of aneuploidy and chromosomal instability. We determined that NUP88 and the nuclear transport factors NUP98 and RAE1 comprise a regulatory network that inhibits premitotic activity of the anaphase-promoting complex/cyclosome (APC/C). When overexpressed, NUP88 sequesters NUP98-RAE1 away from APC/CCDH1, triggering proteolysis of polo-like kinase 1 (PLK1), a tumor suppressor and multitasking mitotic kinase. Premitotic destruction of PLK1 disrupts centrosome separation, causing mitotic spindle asymmetry, merotelic microtubule-kinetochore attachments, lagging chromosomes, and aneuploidy. These effects were replicated by PLK1 insufficiency, indicating that PLK1 is responsible for the mitotic defects associated with NUP88 overexpression. These findings demonstrate that the NUP88-NUP98-RAE1-APC/CCDH1 axis contributes to aneuploidy and suggest that it may be deregulated in the initiating stages of a broad spectrum of human cancers.
Collapse
|
34
|
Basenko EY, Kamei M, Ji L, Schmitz RJ, Lewis ZA. The LSH/DDM1 Homolog MUS-30 Is Required for Genome Stability, but Not for DNA Methylation in Neurospora crassa. PLoS Genet 2016; 12:e1005790. [PMID: 26771905 PMCID: PMC4714748 DOI: 10.1371/journal.pgen.1005790] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 12/16/2015] [Indexed: 01/04/2023] Open
Abstract
LSH/DDM1 enzymes are required for DNA methylation in higher eukaryotes and have poorly defined roles in genome maintenance in yeast, plants, and animals. The filamentous fungus Neurospora crassa is a tractable system that encodes a single LSH/DDM1 homolog (NCU06306). We report that the Neurospora LSH/DDM1 enzyme is encoded by mutagen sensitive-30 (mus-30), a locus identified in a genetic screen over 25 years ago. We show that MUS-30-deficient cells have normal DNA methylation, but are hypersensitive to DNA damaging agents. MUS-30 is a nuclear protein, consistent with its predicted role as a chromatin remodeling enzyme, and levels of MUS-30 are increased following DNA damage. MUS-30 co-purifies with Neurospora WDR76, a homolog of yeast Changed Mutation Rate-1 and mammalian WD40 repeat domain 76. Deletion of wdr76 rescued DNA damage-hypersensitivity of Δmus-30 strains, demonstrating that the MUS-30-WDR76 interaction is functionally important. DNA damage-sensitivity of Δmus-30 is partially suppressed by deletion of methyl adenine glycosylase-1, a component of the base excision repair machinery (BER); however, the rate of BER is not affected in Δmus-30 strains. We found that MUS-30-deficient cells are not defective for DSB repair, and we observed a negative genetic interaction between Δmus-30 and Δmei-3, the Neurospora RAD51 homolog required for homologous recombination. Together, our findings suggest that MUS-30, an LSH/DDM1 homolog, is required to prevent DNA damage arising from toxic base excision repair intermediates. Overall, our study provides important new information about the functions of the LSH/DDM1 family of enzymes.
Collapse
Affiliation(s)
- Evelina Y. Basenko
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Masayuki Kamei
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Lexiang Ji
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
| | - Robert J. Schmitz
- Department of Genetics, University of Georgia, Athens, Georgia, United States of America
| | - Zachary A. Lewis
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
35
|
Sanjiv K, Hagenkort A, Calderón-Montaño JM, Koolmeister T, Reaper PM, Mortusewicz O, Jacques SA, Kuiper RV, Schultz N, Scobie M, Charlton PA, Pollard JR, Berglund UW, Altun M, Helleday T. Cancer-Specific Synthetic Lethality between ATR and CHK1 Kinase Activities. Cell Rep 2015; 14:298-309. [PMID: 26748709 PMCID: PMC4713868 DOI: 10.1016/j.celrep.2015.12.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 11/04/2015] [Accepted: 12/03/2015] [Indexed: 02/05/2023] Open
Abstract
ATR and CHK1 maintain cancer cell survival under replication stress and inhibitors of both kinases are currently undergoing clinical trials. As ATR activity is increased after CHK1 inhibition, we hypothesized that this may indicate an increased reliance on ATR for survival. Indeed, we observe that replication stress induced by the CHK1 inhibitor AZD7762 results in replication catastrophe and apoptosis, when combined with the ATR inhibitor VE-821 specifically in cancer cells. Combined treatment with ATR and CHK1 inhibitors leads to replication fork arrest, ssDNA accumulation, replication collapse, and synergistic cell death in cancer cells in vitro and in vivo. Inhibition of CDK reversed replication stress and synthetic lethality, demonstrating that regulation of origin firing by ATR and CHK1 explains the synthetic lethality. In conclusion, this study exemplifies cancer-specific synthetic lethality between two proteins in the same pathway and raises the prospect of combining ATR and CHK1 inhibitors as promising cancer therapy.
Collapse
Affiliation(s)
- Kumar Sanjiv
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Anna Hagenkort
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - José Manuel Calderón-Montaño
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Tobias Koolmeister
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Philip M Reaper
- Vertex Pharmaceuticals (Europe) Ltd., Abingdon, Oxfordshire OX14 4RW, UK
| | - Oliver Mortusewicz
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Sylvain A Jacques
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Raoul V Kuiper
- Laboratory Medicine, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Niklas Schultz
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Martin Scobie
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Peter A Charlton
- Vertex Pharmaceuticals (Europe) Ltd., Abingdon, Oxfordshire OX14 4RW, UK
| | - John R Pollard
- Vertex Pharmaceuticals (Europe) Ltd., Abingdon, Oxfordshire OX14 4RW, UK
| | - Ulrika Warpman Berglund
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Mikael Altun
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden.
| |
Collapse
|
36
|
Ying S, Chen Z, Medhurst AL, Neal JA, Bao Z, Mortusewicz O, McGouran J, Song X, Shen H, Hamdy FC, Kessler BM, Meek K, Helleday T. DNA-PKcs and PARP1 Bind to Unresected Stalled DNA Replication Forks Where They Recruit XRCC1 to Mediate Repair. Cancer Res 2015; 76:1078-88. [PMID: 26603896 DOI: 10.1158/0008-5472.can-15-0608] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/12/2015] [Indexed: 12/12/2022]
Abstract
A series of critical pathways are responsible for the detection, signaling, and restart of replication forks that encounter blocks during S-phase progression. Small base lesions may obstruct replication fork progression and processing, but the link between repair of small lesions and replication forks is unclear. In this study, we investigated a hypothesized role for DNA-PK, an important enzyme in DNA repair, in cellular responses to DNA replication stress. The enzyme catalytic subunit DNA-PKcs was phosphorylated on S2056 at sites of stalled replication forks in response to short hydroxyurea treatment. Using DNA fiber experiments, we found that catalytically active DNA-PK was required for efficient replication restart of stalled forks. Furthermore, enzymatically active DNA-PK was also required for PARP-dependent recruitment of XRCC1 to stalled replication forks. This activity was enhanced by preventing Mre11-dependent DNA end resection, suggesting that XRCC1 must be recruited early to an unresected stalled fork. We also found that XRCC1 was required for effective restart of a subset of stalled replication forks. Overall, our work suggested that DNA-PK and PARP-dependent recruitment of XRCC1 is necessary to effectively protect, repair, and restart stalled replication forks, providing new insight into how genomic stability is preserved.
Collapse
Affiliation(s)
- Songmin Ying
- Department of Respiratory and Critical Care Medicine of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhihui Chen
- Department of Gastrointestinal and Pancreatic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Annette L Medhurst
- Department of Oncology, CR-UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Jessica A Neal
- Department of Microbiology & Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan. Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Zhengqiang Bao
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Oliver Mortusewicz
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Joanna McGouran
- Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford, United Kingdom
| | - Xinming Song
- Department of Gastrointestinal and Pancreatic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. State Key Laboratory for Respiratory Diseases, Guangzhou, China
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Benedikt M Kessler
- Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford, United Kingdom
| | - Katheryn Meek
- Department of Microbiology & Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan. Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
37
|
Puigvert JC, Sanjiv K, Helleday T. Targeting DNA repair, DNA metabolism and replication stress as anti-cancer strategies. FEBS J 2015; 283:232-45. [DOI: 10.1111/febs.13574] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/04/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Jordi Carreras Puigvert
- Science for Life Laboratory; Division of Translational Medicine and Chemical Biology; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - Kumar Sanjiv
- Science for Life Laboratory; Division of Translational Medicine and Chemical Biology; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - Thomas Helleday
- Science for Life Laboratory; Division of Translational Medicine and Chemical Biology; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
38
|
Wang F, Stewart J, Price CM. Human CST abundance determines recovery from diverse forms of DNA damage and replication stress. Cell Cycle 2015; 13:3488-98. [PMID: 25483097 DOI: 10.4161/15384101.2014.964100] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mammalian CST (CTC1-STN1-TEN1) is a telomere-associated complex that functions in telomere duplex replication and fill-in synthesis of the telomeric C-strand following telomerase action. CST also facilitates genome-wide replication recovery after HU-induced fork stalling by increasing origin firing. CTC1 and STN1 were originally isolated as a DNA polymerase α stimulatory factor. Here we explore how CST abundance affects recovery from drugs that cause different types of DNA damage and replication stress. We show that recovery from HU and aphidicolin induced replication stress is increased by CST over-expression. Elevated CST increases dNTP incorporation and origin firing after HU release and decreases the incidence of anaphase bridges and micronuclei after aphidicolin removal. While the frequency of origin firing after HU release is proportional to CST abundance, the number of cells entering S-phase to initiate replication is unchanged by CST overexpression or STN1 depletion. Instead the CST-related changes in origin firing take place in cells that were already in S-phase at the time of HU addition, indicating that CST modulates firing of late or dormant origins. CST abundance also influences cell viability after treatment with HU, aphidicolin, MMS and camptothecin. Viability is increased by elevated CST and decreased by STN1 depletion, indicating that endogenous CST levels are limiting. However, CST abundance does not affect viability after MMC treatment. Thus, CST facilitates recovery from many, but not all, forms of exogenous DNA damage. Overall our results suggest that CST is needed in stoichiometric amounts to facilitate re-initiation of DNA replication at repaired forks and/or dormant origins.
Collapse
Affiliation(s)
- Feng Wang
- a Department of Cancer Biology ; University of Cincinnati ; Cincinnati , OH USA
| | | | | |
Collapse
|
39
|
Aird KM, Worth AJ, Snyder NW, Lee JV, Sivanand S, Liu Q, Blair IA, Wellen KE, Zhang R. ATM couples replication stress and metabolic reprogramming during cellular senescence. Cell Rep 2015; 11:893-901. [PMID: 25937285 DOI: 10.1016/j.celrep.2015.04.014] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/23/2015] [Accepted: 04/05/2015] [Indexed: 11/18/2022] Open
Abstract
Replication stress induced by nucleotide deficiency plays an important role in cancer initiation. Replication stress in primary cells typically activates the cellular senescence tumor-suppression mechanism. Senescence bypass correlates with development of cancer, a disease characterized by metabolic reprogramming. However, the role of metabolic reprogramming in the cellular response to replication stress has been little explored. Here, we report that ataxia telangiectasia mutated (ATM) plays a central role in regulating the cellular response to replication stress by shifting cellular metabolism. ATM inactivation bypasses senescence induced by replication stress triggered by nucleotide deficiency. This was due to restoration of deoxyribonucleotide triphosphate (dNTP) levels through both upregulation of the pentose phosphate pathway via increased glucose-6-phosphate dehydrogenase (G6PD) activity and enhanced glucose and glutamine consumption. These phenotypes were mediated by a coordinated suppression of p53 and upregulation of c-MYC downstream of ATM inactivation. Our data indicate that ATM status couples replication stress and metabolic reprogramming during senescence.
Collapse
Affiliation(s)
- Katherine M Aird
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew J Worth
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathaniel W Snyder
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joyce V Lee
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sharanya Sivanand
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rugang Zhang
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Kotsantis P, Jones RM, Higgs MR, Petermann E. Cancer therapy and replication stress: forks on the road to perdition. Adv Clin Chem 2015; 69:91-138. [PMID: 25934360 DOI: 10.1016/bs.acc.2014.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deregulated DNA replication occurs in cancer where it contributes to genomic instability. This process is a target of cytotoxic therapies. Chemotherapies exploit high DNA replication in cancer cells by modifying the DNA template or by inhibiting vital enzymatic activities that lead to slowing or stalling replication fork progression. Stalled replication forks can be converted into toxic DNA double-strand breaks resulting in cell death, i.e., replication stress. While likely crucial for many cancer treatments, replication stress is poorly understood due to its complexity. While we still know relatively little about the role of replication stress in cancer therapy, technical advances in recent years have shed new light on the effect that cancer therapeutics have on replication forks and the molecular mechanisms that lead from obstructed fork progression to cell death. This chapter will give an overview of our current understanding of replication stress in the context of cancer therapy.
Collapse
Affiliation(s)
- Panagiotis Kotsantis
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Rebecca M Jones
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Martin R Higgs
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Eva Petermann
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.
| |
Collapse
|
41
|
Truman AW, Kristjansdottir K, Wolfgeher D, Ricco N, Mayampurath A, Volchenboum SL, Clotet J, Kron SJ. Quantitative proteomics of the yeast Hsp70/Hsp90 interactomes during DNA damage reveal chaperone-dependent regulation of ribonucleotide reductase. J Proteomics 2014; 112:285-300. [PMID: 25452130 DOI: 10.1016/j.jprot.2014.09.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 09/05/2014] [Accepted: 09/27/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED The highly conserved molecular chaperones Hsp90 and Hsp70 are indispensible for folding and maturation of a significant fraction of the proteome, including many proteins involved in signal transduction and stress response. To examine the dynamics of chaperone-client interactions after DNA damage, we applied quantitative affinity-purification mass spectrometry (AP-MS) proteomics to characterize interactomes of the yeast Hsp70 isoform Ssa1 and Hsp90 isoform Hsp82 before and after exposure to methyl methanesulfonate. Of 256 proteins identified and quantified via (16)O(/18)O labeling and LC-MS/MS, 142 are novel Hsp70/90 interactors. Nearly all interactions remained unchanged or decreased after DNA damage, but 5 proteins increased interactions with Ssa1 and/or Hsp82, including the ribonucleotide reductase (RNR) subunit Rnr4. Inhibiting Hsp70 or 90 chaperone activity destabilized Rnr4 in yeast and its vertebrate homolog hRMM2 in breast cancer cells. In turn, pre-treatment of cancer cells with chaperone inhibitors sensitized cells to the RNR inhibitor gemcitabine, suggesting a novel chemotherapy strategy. All MS data have been deposited in the ProteomeXchange with identifier PXD001284. BIOLOGICAL SIGNIFICANCE This study provides the dynamic interactome of the yeast Hsp70 and Hsp90 under DNA damage which suggest key roles for the chaperones in a variety of signaling cascades. Importantly, the cancer drug target ribonucleotide reductase was shown to be a client of Hsp70 and Hsp90 in both yeast and breast cancer cells. As such, this study highlights the potential of a novel cancer therapeutic strategy that exploits the synergy of chaperone and ribonucleotide reductase inhibitors.
Collapse
Affiliation(s)
- Andrew W Truman
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | | | - Donald Wolfgeher
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Natalia Ricco
- Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, Barcelona, Catalunya, Spain
| | - Anoop Mayampurath
- Computation Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Samuel L Volchenboum
- Computation Institute, The University of Chicago, Chicago, IL 60637, USA; Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - Josep Clotet
- Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, Barcelona, Catalunya, Spain
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
42
|
Nakamura J, Mutlu E, Sharma V, Collins L, Bodnar W, Yu R, Lai Y, Moeller B, Lu K, Swenberg J. The endogenous exposome. DNA Repair (Amst) 2014; 19:3-13. [PMID: 24767943 PMCID: PMC4097170 DOI: 10.1016/j.dnarep.2014.03.031] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The concept of the Exposome is a compilation of diseases and one's lifetime exposure to chemicals, whether the exposure comes from environmental, dietary, or occupational exposures; or endogenous chemicals that are formed from normal metabolism, inflammation, oxidative stress, lipid peroxidation, infections, and other natural metabolic processes such as alteration of the gut microbiome. In this review, we have focused on the endogenous exposome, the DNA damage that arises from the production of endogenous electrophilic molecules in our cells. It provides quantitative data on endogenous DNA damage and its relationship to mutagenesis, with emphasis on when exogenous chemical exposures that produce identical DNA adducts to those arising from normal metabolism cause significant increases in total identical DNA adducts. We have utilized stable isotope labeled chemical exposures of animals and cells, so that accurate relationships between endogenous and exogenous exposures can be determined. Advances in mass spectrometry have vastly increased both the sensitivity and accuracy of such studies. Furthermore, we have clear evidence of which sources of exposure drive low dose biology that results in mutations and disease. These data provide much needed information to impact quantitative risk assessments, in the hope of moving towards the use of science, rather than default assumptions.
Collapse
Affiliation(s)
- Jun Nakamura
- University of North Carolina, Chapel Hill, NC, United States
| | - Esra Mutlu
- University of North Carolina, Chapel Hill, NC, United States
| | - Vyom Sharma
- University of North Carolina, Chapel Hill, NC, United States
| | - Leonard Collins
- University of North Carolina, Chapel Hill, NC, United States
| | - Wanda Bodnar
- University of North Carolina, Chapel Hill, NC, United States
| | - Rui Yu
- University of North Carolina, Chapel Hill, NC, United States
| | - Yongquan Lai
- University of North Carolina, Chapel Hill, NC, United States
| | - Benjamin Moeller
- University of North Carolina, Chapel Hill, NC, United States; Lovelace Respiratory Research Institute, Albuquerque, NM, United States
| | - Kun Lu
- University of North Carolina, Chapel Hill, NC, United States
| | - James Swenberg
- University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
43
|
Ensminger M, Iloff L, Ebel C, Nikolova T, Kaina B, Lӧbrich M. DNA breaks and chromosomal aberrations arise when replication meets base excision repair. ACTA ACUST UNITED AC 2014; 206:29-43. [PMID: 24982429 PMCID: PMC4085701 DOI: 10.1083/jcb.201312078] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
DNA double-strand breaks and chromosomal aberrations after treatment with N-alkylating agents likely arise as a result of replication fork collision with single-strand breaks generated during base excision repair. Exposures that methylate DNA potently induce DNA double-strand breaks (DSBs) and chromosomal aberrations, which are thought to arise when damaged bases block DNA replication. Here, we demonstrate that DNA methylation damage causes DSB formation when replication interferes with base excision repair (BER), the predominant pathway for repairing methylated bases. We show that cells defective in the N-methylpurine DNA glycosylase, which fail to remove N-methylpurines from DNA and do not initiate BER, display strongly reduced levels of methylation-induced DSBs and chromosomal aberrations compared with wild-type cells. Also, cells unable to generate single-strand breaks (SSBs) at apurinic/apyrimidinic sites do not form DSBs immediately after methylation damage. In contrast, cells deficient in x-ray cross-complementing protein 1, DNA polymerase β, or poly (ADP-ribose) polymerase 1 activity, all of which fail to seal SSBs induced at apurinic/apyrimidinic sites, exhibit strongly elevated levels of methylation-induced DSBs and chromosomal aberrations. We propose that DSBs and chromosomal aberrations after treatment with N-alkylators arise when replication forks collide with SSBs generated during BER.
Collapse
Affiliation(s)
- Michael Ensminger
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Lucie Iloff
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Christian Ebel
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Teodora Nikolova
- Institute of Toxicology, Medical Center of the University Mainz, 55131 Mainz, Germany
| | - Bernd Kaina
- Institute of Toxicology, Medical Center of the University Mainz, 55131 Mainz, Germany
| | - Markus Lӧbrich
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| |
Collapse
|
44
|
Prereplicative repair of oxidized bases in the human genome is mediated by NEIL1 DNA glycosylase together with replication proteins. Proc Natl Acad Sci U S A 2013; 110:E3090-9. [PMID: 23898192 DOI: 10.1073/pnas.1304231110] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Base oxidation by endogenous and environmentally induced reactive oxygen species preferentially occurs in replicating single-stranded templates in mammalian genomes, warranting prereplicative repair of the mutagenic base lesions. It is not clear how such lesions (which, unlike bulky adducts, do not block replication) are recognized for repair. Furthermore, strand breaks caused by base excision from ssDNA by DNA glycosylases, including Nei-like (NEIL) 1, would generate double-strand breaks during replication, which are not experimentally observed. NEIL1, whose deficiency causes a mutator phenotype and is activated during the S phase, is present in the DNA replication complex isolated from human cells, with enhanced association with DNA in S-phase cells and colocalization with replication foci containing DNA replication proteins. Furthermore, NEIL1 binds to 5-hydroxyuracil, the oxidative deamination product of C, in replication protein A-coated ssDNA template and inhibits DNA synthesis by DNA polymerase δ. We postulate that, upon encountering an oxidized base during replication, NEIL1 initiates prereplicative repair by acting as a "cowcatcher" and preventing nascent chain growth. Regression of the stalled replication fork, possibly mediated by annealing helicases, then allows lesion repair in the reannealed duplex. This model is supported by our observations that NEIL1, whose deficiency slows nascent chain growth in oxidatively stressed cells, is stimulated by replication proteins in vitro. Furthermore, deficiency of the closely related NEIL2 alone does not affect chain elongation, but combined NEIL1/2 deficiency further inhibits DNA replication. These results support a mechanism of NEIL1-mediated prereplicative repair of oxidized bases in the replicating strand, with NEIL2 providing a backup function.
Collapse
|
45
|
Abstract
DNA damage created by endogenous or exogenous genotoxic agents can exist in multiple forms, and if allowed to persist, can promote genome instability and directly lead to various human diseases, particularly cancer, neurological abnormalities, immunodeficiency and premature aging. To avoid such deleterious outcomes, cells have evolved an array of DNA repair pathways, which carry out what is typically a multiple-step process to resolve specific DNA lesions and maintain genome integrity. To fully appreciate the biological contributions of the different DNA repair systems, one must keep in mind the cellular context within which they operate. For example, the human body is composed of non-dividing and dividing cell types, including, in the brain, neurons and glial cells. We describe herein the molecular mechanisms of the different DNA repair pathways, and review their roles in non-dividing and dividing cells, with an eye toward how these pathways may regulate the development of neurological disease.
Collapse
Affiliation(s)
- Teruaki Iyama
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | | |
Collapse
|
46
|
Burrell RA, McClelland SE, Endesfelder D, Groth P, Weller MC, Shaikh N, Domingo E, Kanu N, Dewhurst SM, Gronroos E, Chew SK, Rowan AJ, Schenk A, Sheffer M, Howell M, Kschischo M, Behrens A, Helleday T, Bartek J, Tomlinson IP, Swanton C. Replication stress links structural and numerical cancer chromosomal instability. Nature 2013; 494:492-496. [PMID: 23446422 PMCID: PMC4636055 DOI: 10.1038/nature11935] [Citation(s) in RCA: 638] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 01/24/2013] [Indexed: 12/14/2022]
Abstract
Cancer chromosomal instability (CIN) results in an increased rate of change of chromosome number and structure and generates intratumour heterogeneity. CIN is observed in most solid tumours and is associated with both poor prognosis and drug resistance. Understanding a mechanistic basis for CIN is therefore paramount. Here we find evidence for impaired replication fork progression and increased DNA replication stress in CIN(+) colorectal cancer (CRC) cells relative to CIN(-) CRC cells, with structural chromosome abnormalities precipitating chromosome missegregation in mitosis. We identify three new CIN-suppressor genes (PIGN (also known as MCD4), MEX3C (RKHD2) and ZNF516 (KIAA0222)) encoded on chromosome 18q that are subject to frequent copy number loss in CIN(+) CRC. Chromosome 18q loss was temporally associated with aneuploidy onset at the adenoma-carcinoma transition. CIN-suppressor gene silencing leads to DNA replication stress, structural chromosome abnormalities and chromosome missegregation. Supplementing cells with nucleosides, to alleviate replication-associated damage, reduces the frequency of chromosome segregation errors after CIN-suppressor gene silencing, and attenuates segregation errors and DNA damage in CIN(+) cells. These data implicate a central role for replication stress in the generation of structural and numerical CIN, which may inform new therapeutic approaches to limit intratumour heterogeneity.
Collapse
Affiliation(s)
- Rebecca A Burrell
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Sarah E McClelland
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - David Endesfelder
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
- University of Applied Sciences, Mathematics and Techniques, Remagen, Germany
| | - Petra Groth
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Marie-Christine Weller
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Nadeem Shaikh
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Enric Domingo
- Molecular and Population Genetics and NIHR Biomedical Research Centre, The Wellcome Trust Centre for Human Genetics, Oxford, UK
| | - Nnennaya Kanu
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Sally M Dewhurst
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Eva Gronroos
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Su Kit Chew
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
- UCL Cancer Institute, Paul O'Gorman Building, Huntley St., London, UK
| | - Andrew J Rowan
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Arne Schenk
- University of Applied Sciences, Mathematics and Techniques, Remagen, Germany
| | - Michal Sheffer
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Howell
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Maik Kschischo
- University of Applied Sciences, Mathematics and Techniques, Remagen, Germany
| | - Axel Behrens
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jiri Bartek
- Danish Cancer Society Research Center, Strandboulevarden 49, Copenhagen, Denmark
- Institute of Molecular and Translational Medicine, Palacky University Olomouc, Czech republic
| | - Ian P Tomlinson
- Molecular and Population Genetics and NIHR Biomedical Research Centre, The Wellcome Trust Centre for Human Genetics, Oxford, UK
| | - Charles Swanton
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, UK
- UCL Cancer Institute, Paul O'Gorman Building, Huntley St., London, UK
| |
Collapse
|
47
|
Strategies for the Use of Poly(adenosine diphosphate ribose) Polymerase (PARP) Inhibitors in Cancer Therapy. Biomolecules 2012; 2:635-49. [PMID: 24970153 PMCID: PMC4030856 DOI: 10.3390/biom2040635] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 11/29/2012] [Accepted: 12/09/2012] [Indexed: 11/16/2022] Open
Abstract
Treatments with Poly(adenosine diphosphate ribose) polymerase (PARP) inhibitors have offered patients carrying cancers with mutated BRCA1 or BRCA2 genes a new and in many cases effective option for disease control. There is potentially a large patient population that may also benefit from PARP inhibitor treatment, either in monotherapy or in combination with chemotherapy. Here, we describe the multifaceted role of PARP inhibitors and discuss which treatment options could potentially be useful to gain disease control without potentiating side effects.
Collapse
|
48
|
Vare D, Groth P, Carlsson R, Johansson F, Erixon K, Jenssen D. DNA interstrand crosslinks induce a potent replication block followed by formation and repair of double strand breaks in intact mammalian cells. DNA Repair (Amst) 2012; 11:976-85. [DOI: 10.1016/j.dnarep.2012.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 09/17/2012] [Accepted: 09/19/2012] [Indexed: 11/17/2022]
|
49
|
Fenske AE, Glaesener S, Bokemeyer C, Thomale J, Dahm-Daphi J, Honecker F, Dartsch DC. Cisplatin resistance induced in germ cell tumour cells is due to reduced susceptibility towards cell death but not to altered DNA damage induction or repair. Cancer Lett 2012; 324:171-8. [DOI: 10.1016/j.canlet.2012.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 05/09/2012] [Accepted: 05/11/2012] [Indexed: 11/26/2022]
|
50
|
Tittel-Elmer M, Lengronne A, Davidson MB, Bacal J, François P, Hohl M, Petrini JHJ, Pasero P, Cobb JA. Cohesin association to replication sites depends on rad50 and promotes fork restart. Mol Cell 2012; 48:98-108. [PMID: 22885006 PMCID: PMC3904740 DOI: 10.1016/j.molcel.2012.07.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 05/21/2012] [Accepted: 07/05/2012] [Indexed: 01/19/2023]
Abstract
The cohesin complex holds together newly replicated chromatids and is involved in diverse pathways that preserve genome integrity. We show that in budding yeast, cohesin is transiently recruited to active replication origins, and it spreads along DNA as forks progress. When DNA synthesis is impeded, cohesin accumulates at replication sites and is critical for the recovery of stalled forks. Cohesin enrichment at replication forks does not depend on γH2A(X) formation, which differs from its loading requirements at DNA double-strand breaks (DSBs). However, cohesin localization is largely reduced in rad50Δ mutants and in cells lacking both Mec1 and Tel1 checkpoint kinases. Interestingly, cohesin loading at replication sites depends on the structural features of Rad50 that are important for bridging sister chromatids, including the CXXC hook domain and the length of the coiled-coil extensions. Together, these data reveal a function for cohesin in the maintenance of genome integrity during S phase.
Collapse
Affiliation(s)
- Mireille Tittel-Elmer
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB Canada T2N 4N1
- Institute of Human Genetics, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1142, Montpellier, France
| | - Armelle Lengronne
- Institute of Human Genetics, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1142, Montpellier, France
| | - Marta B Davidson
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB Canada T2N 4N1
| | - Julien Bacal
- Institute of Human Genetics, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1142, Montpellier, France
| | - Philippe François
- Institute of Human Genetics, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1142, Montpellier, France
| | - Marcel Hohl
- Laboratory of Chromosome Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - John H J Petrini
- Laboratory of Chromosome Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Philippe Pasero
- Institute of Human Genetics, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1142, Montpellier, France
| | - Jennifer A Cobb
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB Canada T2N 4N1
| |
Collapse
|