1
|
Icsel C, Aydinlik S, Aygun M, Yilmaz VT. Cationic first-row transition metal saccharinate complexes with tris(2-pyridylmethyl)amine: synthesis, structures and anticancer studies. Dalton Trans 2024; 54:247-258. [PMID: 39530863 DOI: 10.1039/d4dt02258k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
A series of new cationic first-row transition metal complexes of [Mn(sac)(H2O)(tpma)](sac)·H2O (Mn), [(μ-O){FeCl(tpma)}2](sac)2·3H2O (Fe), [Co(sac)(H2O)(tpma)](sac)·H2O (Co), [Ni(H2O)2(tpma)](sac)2·2H2O (Ni), [Cu(sac)(tpma)](sac) (Cu) and [Zn(sac)(H2O)(tpma)](sac) (Zn), where sac = saccharinate and tpma = tris(2-pyridylmethyl)amine, were synthesized and structurally characterized by elemental analysis, UV-Vis, IR, ESI-MS, NMR, X-ray diffraction and conductivity measurements. The cytotoxic activity of the metal complexes was evaluated in vitro against lung carcinoma (A549), breast adenocarcinoma (MCF7), colon (HT29), and normal BEAS-2B cell lines. Mn and Fe displayed potent cytotoxic activity in all cell lines with IC50 values between 1.99 ± 0.33 and 6.65 ± 0.67 μM, while Cu moderately affected the growth of HT29 cells. However the rest of the metal complexes did not demonstrate any growth inhibitory effect. Further studies with Fe treated HT29 cells through cellular imaging analysis indicated that Fe significantly induced intracellular ROS (reactive oxygen species) accumulation, mitochondrial dysfunction and double-strand DNA breaks, and eventually caused apoptotic cell death through the intrinsic pathway.
Collapse
Affiliation(s)
- Ceyda Icsel
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, 61080 Trabzon, Turkey.
| | - Seyma Aydinlik
- TUBITAK Marmara Research Center, Climate and Life Sciences, Biotechnology Research Group, 41470 Gebze, Kocaeli, Turkey
| | - Muhittin Aygun
- Department of Physics, Faculty of Sciences, Dokuz Eylul University, 35210 Izmir, Turkey
| | - Veysel T Yilmaz
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, 61080 Trabzon, Turkey.
| |
Collapse
|
2
|
Diaz-Diaz S, Recacha E, Pulido MR, Romero-Muñoz M, de Gregorio-Iaria B, Docobo-Pérez F, Pascual A, Rodríguez-Martínez JM. Synergistic Effect of SOS Response and GATC Methylome Suppression on Antibiotic Stress Survival in Escherichia coli. Antimicrob Agents Chemother 2023; 67:e0139222. [PMID: 36802234 PMCID: PMC10019295 DOI: 10.1128/aac.01392-22] [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: 10/14/2022] [Accepted: 01/22/2023] [Indexed: 02/23/2023] Open
Abstract
The suppression of the SOS response has been shown to enhance the in vitro activity of quinolones. Furthermore, Dam-dependent base methylation has an impact on susceptibility to other antimicrobials affecting DNA synthesis. Here, we investigated the interplay between these two processes, alone and in combination, in terms of antimicrobial activity. A genetic strategy was used employing single- and double-gene mutants for the SOS response (recA gene) and the Dam methylation system (dam gene) in isogenic models of Escherichia coli both susceptible and resistant to quinolones. Regarding the bacteriostatic activity of quinolones, a synergistic sensitization effect was observed when the Dam methylation system and the recA gene were suppressed. In terms of growth, after 24 h in the presence of quinolones, the Δdam ΔrecA double mutant showed no growth or delayed growth compared to the control strain. In bactericidal terms, spot tests showed that the Δdam ΔrecA double mutant was more sensitive than the ΔrecA single mutant (about 10- to 102-fold) and the wild type (about 103- to 104-fold) in both susceptible and resistant genetic backgrounds. Differences between the wild type and the Δdam ΔrecA double mutant were confirmed by time-kill assays. The suppression of both systems, in a strain with chromosomal mechanisms of quinolone resistance, prevents the evolution of resistance. This genetic and microbiological approach demonstrated the enhanced sensitization of E. coli to quinolones by dual targeting of the recA (SOS response) and Dam methylation system genes, even in a resistant strain model.
Collapse
Affiliation(s)
- S. Diaz-Diaz
- Unidad de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Seville, Spain
| | - E. Recacha
- Unidad de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Seville, Spain
| | - Marina R. Pulido
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Seville, Spain
| | - María Romero-Muñoz
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - B. de Gregorio-Iaria
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - F. Docobo-Pérez
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Seville, Spain
| | - A. Pascual
- Unidad de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Seville, Spain
| | - J. M. Rodríguez-Martínez
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Seville, Spain
| |
Collapse
|
3
|
McKenzie AM, Henry C, Myers KS, Place MM, Keck JL. Identification of genetic interactions with priB links the PriA/PriB DNA replication restart pathway to double-strand DNA break repair in Escherichia coli. G3 (BETHESDA, MD.) 2022; 12:jkac295. [PMID: 36326440 PMCID: PMC9713433 DOI: 10.1093/g3journal/jkac295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/20/2022] [Indexed: 11/30/2023]
Abstract
Collisions between DNA replication complexes (replisomes) and impediments such as damaged DNA or proteins tightly bound to the chromosome lead to premature dissociation of replisomes at least once per cell cycle in Escherichia coli. Left unrepaired, these events produce incompletely replicated chromosomes that cannot be properly partitioned into daughter cells. DNA replication restart, the process that reloads replisomes at prematurely terminated sites, is therefore essential in E. coli and other bacteria. Three replication restart pathways have been identified in E. coli: PriA/PriB, PriA/PriC, and PriC/Rep. A limited number of genetic interactions between replication restart and other genome maintenance pathways have been defined, but a systematic study placing replication restart reactions in a broader cellular context has not been performed. We have utilized transposon-insertion sequencing to identify new genetic interactions between DNA replication restart pathways and other cellular systems. Known genetic interactors with the priB replication restart gene (uniquely involved in the PriA/PriB pathway) were confirmed and several novel priB interactions were discovered. Targeted genetic and imaging-based experiments with priB and its genetic partners revealed significant double-strand DNA break accumulation in strains with mutations in dam, rep, rdgC, lexA, or polA. Modulating the activity of the RecA recombinase partially suppressed the detrimental effects of rdgC or lexA mutations in ΔpriB cells. Taken together, our results highlight roles for several genes in double-strand DNA break homeostasis and define a genetic network that facilitates DNA repair/processing upstream of PriA/PriB-mediated DNA replication restart in E. coli.
Collapse
Affiliation(s)
- Aidan M McKenzie
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Camille Henry
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kevin S Myers
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Michael M Place
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - James L Keck
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| |
Collapse
|
4
|
Panzarino NJ, Krais JJ, Cong K, Peng M, Mosqueda M, Nayak SU, Bond SM, Calvo JA, Doshi MB, Bere M, Ou J, Deng B, Zhu LJ, Johnson N, Cantor SB. Replication Gaps Underlie BRCA Deficiency and Therapy Response. Cancer Res 2021; 81:1388-1397. [PMID: 33184108 PMCID: PMC8026497 DOI: 10.1158/0008-5472.can-20-1602] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/02/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
Defects in DNA repair and the protection of stalled DNA replication forks are thought to underlie the chemosensitivity of tumors deficient in the hereditary breast cancer genes BRCA1 and BRCA2 (BRCA). Challenging this assumption are recent findings that indicate chemotherapies, such as cisplatin used to treat BRCA-deficient tumors, do not initially cause DNA double-strand breaks (DSB). Here, we show that ssDNA replication gaps underlie the hypersensitivity of BRCA-deficient cancer and that defects in homologous recombination (HR) or fork protection (FP) do not. In BRCA-deficient cells, ssDNA gaps developed because replication was not effectively restrained in response to stress. Gap suppression by either restoration of fork restraint or gap filling conferred therapy resistance in tissue culture and BRCA patient tumors. In contrast, restored FP and HR could be uncoupled from therapy resistance when gaps were present. Moreover, DSBs were not detected after therapy when apoptosis was inhibited, supporting a framework in which DSBs are not directly induced by genotoxic agents, but rather are induced from cell death nucleases and are not fundamental to the mechanism of action of genotoxic agents. Together, these data indicate that ssDNA replication gaps underlie the BRCA cancer phenotype, "BRCAness," and we propose they are fundamental to the mechanism of action of genotoxic chemotherapies. SIGNIFICANCE: This study suggests that ssDNA replication gaps are fundamental to the toxicity of genotoxic agents and underlie the BRCA-cancer phenotype "BRCAness," yielding promising biomarkers, targets, and opportunities to resensitize refractory disease.See related commentary by Canman, p. 1214.
Collapse
Affiliation(s)
| | - John J Krais
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ke Cong
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Min Peng
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michelle Mosqueda
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Sumeet U Nayak
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Samuel M Bond
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jennifer A Calvo
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Mihir B Doshi
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Matt Bere
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jianhong Ou
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Bin Deng
- The University of Vermont, Burlington, Vermont
| | - Lihua J Zhu
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Neil Johnson
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sharon B Cantor
- University of Massachusetts Medical School, Worcester, Massachusetts.
| |
Collapse
|
5
|
Lettl C, Schindele F, Testolin G, Bär A, Rehm T, Brönstrup M, Schobert R, Bilitewski U, Haas R, Fischer W. Inhibition of Type IV Secretion Activity and Growth of Helicobacter pylori by Cisplatin and Other Platinum Complexes. Front Cell Infect Microbiol 2020; 10:602958. [PMID: 33392108 PMCID: PMC7775389 DOI: 10.3389/fcimb.2020.602958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
Type IV secretion systems are protein secretion machineries that are frequently used by pathogenic bacteria to inject their virulence factors into target cells of their respective hosts. In the case of the human gastric pathogen Helicobacter pylori, the cytotoxin-associated gene (Cag) type IV secretion system is considered a major cause for severe disease, such as gastric cancer, and thus constitutes an attractive target for specific treatment options against H. pylori infections. Here, we have used a Cag type IV secretion reporter assay for screening a repurposing compound library for inhibitors targeting this system. We found that the antitumor agent cisplatin, a platinum coordination complex that kills target cells by formation of DNA crosslinks, is a potent inhibitor of the Cag type IV secretion system. Strikingly, we found that this inhibitory activity of cisplatin depends on a ligand exchange reaction which incorporates a solvent molecule (dimethylsulfoxide) into the complex, a modification which is known to be deleterious for DNA crosslinking, and for its anticancer activity. We extended our analysis to several analogous platinum complexes containing N-heterocyclic carbene, as well as DMSO or other ligands, and found varying inhibitory activities toward the Cag system which were not congruent with their DNA-binding properties, suggesting that protein interactions may cause the inhibitory effect. Inhibition experiments under varying conditions revealed effects on adherence and bacterial viability as well, and showed that the type IV secretion-inhibitory capacity of platinum complexes can be inactivated by sulfur-containing reagents and in complex bacterial growth media. Taken together, our results demonstrate DNA binding-independent inhibitory effects of cisplatin and other platinum complexes against different H. pylori processes including type IV secretion.
Collapse
Affiliation(s)
- Clara Lettl
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| | - Franziska Schindele
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| | - Giambattista Testolin
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Alexander Bär
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth, Germany
| | - Tobias Rehm
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth, Germany
| | - Ursula Bilitewski
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Rainer Haas
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| | - Wolfgang Fischer
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| |
Collapse
|
6
|
Bouberhan S, Philp L, Hill S, Al-Alem LF, Rueda B. Exploiting the Prevalence of Homologous Recombination Deficiencies in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2020; 12:E1206. [PMID: 32403357 PMCID: PMC7281458 DOI: 10.3390/cancers12051206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 01/07/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) remains the most lethal gynecologic cancer in the United States. Genomic analysis revealed roughly half of HGSOC display homologous repair deficiencies. An improved understanding of the genomic and somatic mutations that influence DNA repair led to the development of poly(ADP-ribose) polymerase inhibitors for the treatment of ovarian cancer. In this review, we explore the preclinical and clinical studies that led to the development of FDA approved drugs that take advantage of the synthetic lethality concept, the implementation of the early phase trials, the development of companion diagnostics and proposed mechanisms of resistance.
Collapse
Affiliation(s)
- Sara Bouberhan
- Department of Hematology/Medical Oncology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Department of Hematology/Medical Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren Philp
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Hill
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA;
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Linah F. Al-Alem
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bo Rueda
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Huang HK, Lin YH, Chang HA, Lai YS, Chen YC, Huang SC, Chou CY, Chiu WT. Chemoresistant ovarian cancer enhances its migration abilities by increasing store-operated Ca 2+ entry-mediated turnover of focal adhesions. J Biomed Sci 2020; 27:36. [PMID: 32079527 PMCID: PMC7033940 DOI: 10.1186/s12929-020-00630-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/17/2020] [Indexed: 01/05/2023] Open
Abstract
Background Among gynecological cancers, ovarian carcinoma has the highest mortality rate, and chemoresistance is highly prevalent in this cancer. Therefore, novel strategies are required to improve its poor prognosis. Formation and disassembly of focal adhesions are regulated dynamically during cell migration, which plays an essential role in cancer metastasis. Metastasis is intricately linked with resistance to chemotherapy, but the molecular basis for this link is unknown. Methods Transwell migration and wound healing migration assays were used to analyze the migration ability of ovarian cancer cells. Real-time recordings by total internal reflection fluorescence microscope (TIRFM) were performed to assess the turnover of focal adhesions with fluorescence protein-tagged focal adhesion molecules. SOCE inhibitors were used to verify the effects of SOCE on focal adhesion dynamics, cell migration, and chemoresistance in chemoresistant cells. Results We found that mesenchymal-like chemoresistant IGROV1 ovarian cancer cells have higher migration properties because of their rapid regulation of focal adhesion dynamics through FAK, paxillin, vinculin, and talin. Focal adhesions in chemoresistant cells, they were smaller and exhibited strong adhesive force, which caused the cells to migrate rapidly. Store-operated Ca2+ entry (SOCE) regulates focal adhesion turnover, and cell polarization and migration. Herein, we compared SOCE upregulation in chemoresistant ovarian cancer cells to its parental cells. SOCE inhibitors attenuated the assembly and disassembly of focal adhesions significantly. Results of wound healing and transwell assays revealed that SOCE inhibitors decreased chemoresistant cell migration. Additionally, SOCE inhibitors combined with chemotherapeutic drugs could reverse ovarian cancer drug resistance. Conclusion Our findings describe the role of SOCE in chemoresistance-mediated focal adhesion turnover, cell migration, and viability. Consequently, SOCE might be a promising therapeutic target in epithelial ovarian cancer. Graphical abstract ![]()
Collapse
Affiliation(s)
- Ho-Kai Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yi-Hsin Lin
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Heng-Ai Chang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yi-Shyun Lai
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ying-Chi Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Soon-Cen Huang
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Liouying Campus, Tainan, 736, Taiwan
| | - Cheng-Yang Chou
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan. .,Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, 701, Taiwan. .,Medical Device Innovation Center, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
8
|
Liu Z, Yu M, Fei B, Sun J, Wang D. Identification Of Natural Compound Derivative For Inhibition Of XLF And Overcoming Chemoresistance In Colorectal Cancer Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3823-3834. [PMID: 31806933 PMCID: PMC6847993 DOI: 10.2147/dddt.s215967] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Abstract
Purpose A previous study has identified that XRCC4-like factor (XLF) is a potential target to overcome resistance to 5-fluorouracil (5-Fu) and oxaliplatin (OXA) in colorectal cancer (CRC). The purpose of this study is to develop potent XLF inhibitors to chemoresistance in CRC. Methods Virtual screening was adopted to identify novel XLF-binding compounds by initially testing 6800 molecules in Chemical Entities of Biological Interest library. Hit compounds were further validated by Western blot assay. Cell sensitivity to 5-Fu and OXA was measured using sulforhodamine B assay. The effect of XLF inhibitor on DNA repair efficiency was evaluated by comet assay, fluorescent-based nonhomologous end joining (NHEJ) and homologous recombination (HR) reporter assays. DNA-binding activity of NHEJ key factors was examined by chromatin fractionation assay. Results We identified G3, a novel and potent XLF inhibitor (IC50 0.47±0.02 µM). G3 induced XLF protein degradation in CRC cells. Significantly, G3 improved cell sensitivity to 5-Fu and OXA in chemoresistant CRC cell lines. Mechanistically, G3 depleted XLF expression, severely compromised NHEJ efficiency by up to 65% and inhibited NHEJ key factor assembly on DNA. G3 also inhibited HR efficiency in a time-dependent manner. Conclusion These results suggest that G3 overcomes 5-Fu and OXA resistance in CRC cells by inhibiting XLF expression. Thus, XLF is a promising target and its inhibitor G3 is a potential candidate for treatment of chemoresistant CRC patients.
Collapse
Affiliation(s)
- Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Miao Yu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Bingyuan Fei
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jing Sun
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Dongxin Wang
- Department of Anesthesiology, Jilin Cancer Hospital, Jilin, People's Republic of China
| |
Collapse
|
9
|
Oliveira L, Caquito Jr JM, Rocha MS. Oxaliplatin effects on the DNA molecule studied by force spectroscopy. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab37ce] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
10
|
Rahman FU, Ali A, Khan IU, Bhatti MZ, Petroselli M, Duong HQ, Martì-Rujas J, Li ZT, Wang H, Zhang DW. Monofunctional supramolecular Pt(II) complexes: Synthesis, single crystal structure, anticancer activity, E. coli growth retardation and DNA interaction study. INORG CHEM COMMUN 2019. [DOI: 10.1016/j.inoche.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Oliveira L, Caquito J, Rocha M. Carboplatin as an alternative to Cisplatin in chemotherapies: New insights at single molecule level. Biophys Chem 2018; 241:8-14. [DOI: 10.1016/j.bpc.2018.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 07/20/2018] [Indexed: 10/28/2022]
|
12
|
Xie J, Gurler Main H, Sacks JD, Muralidhar GG, Barbolina MV. Regulation of DNA damage repair and lipid uptake by CX 3CR1 in epithelial ovarian carcinoma. Oncogenesis 2018; 7:37. [PMID: 29712888 PMCID: PMC5928120 DOI: 10.1038/s41389-018-0046-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/17/2018] [Accepted: 03/26/2018] [Indexed: 12/25/2022] Open
Abstract
Failure of currently used cytotoxic chemotherapy is one of the main reasons behind high mortality from metastatic high grade serous ovarian carcinoma. We found that high expression of a receptor for fractalkine (CX3CR1) significantly correlated with shorter survival of patients with serous ovarian carcinoma treated with cytotoxic DNA damage chemotherapies, and reduction of CX3CR1 expression resulted in sensitization to several DNA damaging modalities, including x-ray radiation and cisplatin. Here, we show that CX3CR1 plays a role in double-strand DNA break response and repair by regulating expression of RAD50 by a MYC-dependent mechanism. We demonstrate that downregulation of CX3CR1 alone and in a combination with irradiation affects peritoneal metastasis in an organ-specific manner; we show that CX3CR1 regulates lipid uptake which could control omental metastasis. This study identifies CX3CR1 as a novel potential target for sensitization of ovarian carcinoma to DNA damage therapies and reduction of peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Jia Xie
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Hilal Gurler Main
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Joelle D Sacks
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Goda G Muralidhar
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Maria V Barbolina
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
13
|
Wang X, Gu Y, Liu H, Shi L, Sun X. Icotinib hydrochloride enhances chemo- and radiosensitivity by inhibiting EGFR signaling and attenuating RAD51 expression and function in Hela S3 cells. Onco Targets Ther 2018; 11:1245-1258. [PMID: 29551903 PMCID: PMC5843137 DOI: 10.2147/ott.s152613] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Radiotherapy and cisplatin-based chemotherapy are currently considered as standard treatments employed for advanced cervical cancer (CC). However, patients with local recurrence or distant metastasis continue to have poor outcomes. EGFR overexpression correlated with chemo/radioresistance, and disease failure has been well proved in the previous studies. Hence, the aim of this study was to explore the therapeutic efficacy and underlying mechanism of the sensitization to radiation or cisplatin of icotinib hydrochloride (IH), a high-selective EGFR tyrosine kinase inhibitor (TKI), in the Hela S3 human CC cell line. Methods Cell proliferation was measured with cell counting kit-8 (CCK-8) assay. Flow cytometry analysis was performed to examine cell cycle distribution and apoptosis. The phosphorylation of EGFR and its downstream signaling molecules were measured by Western blot analysis. γ-H2AX foci and RAD51 foci in the cellular nucleus were visualized using immunofluoresence staining. Expression levels of RAD51 in the whole cells and subceullar fractions were detected to demonstrate the impact of IH on DNA repair. Results IH can significantly inhibit cell proliferation, redistribute cell cycle, enhance apoptosis and impair DNA damage response of Hela S3 cells following radiation or cisplatin treatment through suppressing the activation of the EGFR signaling pathway and attenuating the expression and function of homologous recombination (HR) protein RAD51. Conclusion This study suggests that IH is a potential sensitizer in radiotherapy and cisplatin-based chemotherapy for CC and RAD51 may serve as a prognosis biomarker for this combination treatment.
Collapse
Affiliation(s)
- Xuanxuan Wang
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanjun Gu
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hai Liu
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liming Shi
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaonan Sun
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Windgassen TA, Wessel SR, Bhattacharyya B, Keck JL. Mechanisms of bacterial DNA replication restart. Nucleic Acids Res 2018; 46:504-519. [PMID: 29202195 PMCID: PMC5778457 DOI: 10.1093/nar/gkx1203] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/15/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022] Open
Abstract
Multi-protein DNA replication complexes called replisomes perform the essential process of copying cellular genetic information prior to cell division. Under ideal conditions, replisomes dissociate only after the entire genome has been duplicated. However, DNA replication rarely occurs without interruptions that can dislodge replisomes from DNA. Such events produce incompletely replicated chromosomes that, if left unrepaired, prevent the segregation of full genomes to daughter cells. To mitigate this threat, cells have evolved 'DNA replication restart' pathways that have been best defined in bacteria. Replication restart requires recognition and remodeling of abandoned replication forks by DNA replication restart proteins followed by reloading of the replicative DNA helicase, which subsequently directs assembly of the remaining replisome subunits. This review summarizes our current understanding of the mechanisms underlying replication restart and the proteins that drive the process in Escherichia coli (PriA, PriB, PriC and DnaT).
Collapse
Affiliation(s)
- Tricia A Windgassen
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Sarah R Wessel
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
- Department of Biochemistry, Vanderbilt School of Medicine, Nashville, TN 37205, USA
| | - Basudeb Bhattacharyya
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
- Department of Chemistry and Biochemistry, University of Wisconsin-La Crosse, La Crosse, WI 54601, USA
| | - James L Keck
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| |
Collapse
|
15
|
Oliveira L, Rocha MS. Force spectroscopy unravels the role of ionic strength on DNA-cisplatin interaction: Modulating the binding parameters. Phys Rev E 2017; 96:032408. [PMID: 29346883 DOI: 10.1103/physreve.96.032408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Indexed: 06/07/2023]
Abstract
In the present work we have gone a step forward in the understanding of the DNA-cisplatin interaction, investigating the role of the ionic strength on the complexes formation. To achieve this task, we use optical tweezers to perform force spectroscopy on the DNA-cisplatin complexes, determining their mechanical parameters as a function of the drug concentration in the sample for three different buffers. From such measurements, we determine the binding parameters and study their behavior as a function of the ionic strength. The equilibrium binding constant decreases with the counterion concentration ([Na]) and can be used to estimate the effective net charge of cisplatin in solution. The cooperativity degree of the binding reaction, on the other hand, increases with the ionic strength, as a result of the different conformational changes induced by the drug on the double-helix when binding under different buffer conditions. Such results can be used to modulate the drug binding to DNA, by appropriately setting the ionic strength of the surrounding buffer. The conclusions drawn provide significant new insights on the complex cooperative interactions between the DNA molecule and the class of platinum-based compounds, much used in chemotherapies.
Collapse
Affiliation(s)
- L Oliveira
- Laboratório de Física Biológica, Departamento de Física, Universidade Federal de Viçosa. Viçosa, Minas Gerais, Brazil
| | - M S Rocha
- Laboratório de Física Biológica, Departamento de Física, Universidade Federal de Viçosa. Viçosa, Minas Gerais, Brazil
| |
Collapse
|
16
|
Abstract
The DNA of Escherichia coli contains 19,120 6-methyladenines and 12,045 5-methylcytosines in addition to the four regular bases, and these are formed by the postreplicative action of three DNA methyltransferases. The majority of the methylated bases are formed by the Dam and Dcm methyltransferases encoded by the dam (DNA adenine methyltransferase) and dcm (DNA cytosine methyltransferase) genes. Although not essential, Dam methylation is important for strand discrimination during the repair of replication errors, controlling the frequency of initiation of chromosome replication at oriC, and the regulation of transcription initiation at promoters containing GATC sequences. In contrast, there is no known function for Dcm methylation, although Dcm recognition sites constitute sequence motifs for Very Short Patch repair of T/G base mismatches. In certain bacteria (e.g., Vibrio cholerae, Caulobacter crescentus) adenine methylation is essential, and, in C. crescentus, it is important for temporal gene expression, which, in turn, is required for coordinating chromosome initiation, replication, and division. In practical terms, Dam and Dcm methylation can inhibit restriction enzyme cleavage, decrease transformation frequency in certain bacteria, and decrease the stability of short direct repeats and are necessary for site-directed mutagenesis and to probe eukaryotic structure and function.
Collapse
|
17
|
Rahman FU, Ali A, Khan I, Guo R, Chen L, Wang H, Li ZT, Lin Y, Zhang DW. Synthesis and characterization of trans-Pt(II)(salicylaldimine)(pyridine/pyridine-4-carbinol)Cl complexes: In vivo inhibition of E. coli growth and in vitro anticancer activities. Polyhedron 2015. [DOI: 10.1016/j.poly.2015.08.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
18
|
Pharmacologic inhibition of ATR and ATM offers clinically important distinctions to enhancing platinum or radiation response in ovarian, endometrial, and cervical cancer cells. Gynecol Oncol 2015; 136:554-61. [PMID: 25560806 DOI: 10.1016/j.ygyno.2014.12.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/17/2014] [Accepted: 12/25/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Significant reductions in gynecologic (GYN) cancer mortality and morbidity require treatments that prevent and reverse resistance to chemotherapy and radiation. The objective of this study was to determine if pharmacologic inhibition of key DNA damage response kinases in GYN cancers would enhance cell killing by platinum-based chemotherapy and radiation. METHODS A panel of human ovarian, endometrial and cervical cancer cell lines were treated with platinum drugs or ionizing radiation (IR) along with small molecule pharmacological kinase inhibitors of Ataxia telangiectasia mutated (ATM) and ATM and Rad-3-related (ATR). RESULTS Pharmacologic inhibition of ATR significantly enhanced platinum drug response in all GYN cancer cell lines tested, whereas inhibition of ATM did not enhance the response to platinum drugs. Co-inhibition of ATM and ATR did not enhance platinum kill beyond that observed by inhibition of ATR alone. By contrast, inhibiting either ATR or ATM enhanced the response to IR in all GYN cancer cells, with further enhancement achieved with co-inhibition. CONCLUSIONS These studies highlight actionable mechanisms operative in GYN cancer cells with potential to maximize response of platinum agents and radiation in newly diagnosed as well as recurrent gynecologic cancers.
Collapse
|
19
|
Peng M, Xie J, Ucher A, Stavnezer J, Cantor SB. Crosstalk between BRCA-Fanconi anemia and mismatch repair pathways prevents MSH2-dependent aberrant DNA damage responses. EMBO J 2014; 33:1698-712. [PMID: 24966277 PMCID: PMC4194102 DOI: 10.15252/embj.201387530] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Several proteins in the BRCA-Fanconi anemia (FA) pathway, such as FANCJ, BRCA1, and FANCD2, interact with mismatch repair (MMR) pathway factors, but the significance of this link remains unknown. Unlike the BRCA-FA pathway, the MMR pathway is not essential for cells to survive toxic DNA interstrand crosslinks (ICLs), although MMR proteins bind ICLs and other DNA structures that form at stalled replication forks. We hypothesized that MMR proteins corrupt ICL repair in cells that lack crosstalk between BRCA-FA and MMR pathways. Here, we show that ICL sensitivity of cells lacking the interaction between FANCJ and the MMR protein MLH1 is suppressed by depletion of the upstream mismatch recognition factor MSH2. MSH2 depletion suppresses an aberrant DNA damage response, restores cell cycle progression, and promotes ICL resistance through a Rad18-dependent mechanism. MSH2 depletion also suppresses ICL sensitivity in cells deficient for BRCA1 or FANCD2, but not FANCA. Rescue by Msh2 loss was confirmed in Fancd2-null primary mouse cells. Thus, we propose that regulation of MSH2-dependent DNA damage response underlies the importance of interactions between BRCA-FA and MMR pathways.
Collapse
Affiliation(s)
- Min Peng
- Department of Cancer Biology, University of Massachusetts Medical School, Women's Cancers Program, UMASS Memorial Cancer Center, Worcester, MA, USA
| | - Jenny Xie
- Department of Cancer Biology, University of Massachusetts Medical School, Women's Cancers Program, UMASS Memorial Cancer Center, Worcester, MA, USA
| | - Anna Ucher
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Women's Cancers Program, UMASS Memorial Cancer Center, Worcester, MA, USA
| | - Janet Stavnezer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Women's Cancers Program, UMASS Memorial Cancer Center, Worcester, MA, USA
| | - Sharon B Cantor
- Department of Cancer Biology, University of Massachusetts Medical School, Women's Cancers Program, UMASS Memorial Cancer Center, Worcester, MA, USA
| |
Collapse
|
20
|
Val ME, Kennedy SP, Soler-Bistué AJ, Barbe V, Bouchier C, Ducos-Galand M, Skovgaard O, Mazel D. Fuse or die: how to survive the loss of Dam inVibrio cholerae. Mol Microbiol 2014; 91:665-78. [DOI: 10.1111/mmi.12483] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2013] [Indexed: 11/26/2022]
Affiliation(s)
- Marie-Eve Val
- Department of Genomes and Genetics; Institut Pasteur; F-75015 Paris France
- CNRS; UMR3525 F-75015 Paris France
| | | | - Alfonso J. Soler-Bistué
- Department of Genomes and Genetics; Institut Pasteur; F-75015 Paris France
- CNRS; UMR3525 F-75015 Paris France
| | | | | | - Magaly Ducos-Galand
- Department of Genomes and Genetics; Institut Pasteur; F-75015 Paris France
- CNRS; UMR3525 F-75015 Paris France
| | - Ole Skovgaard
- Department of Science, Systems and Models; Roskilde University; DK-4000 Roskilde Denmark
| | - Didier Mazel
- Department of Genomes and Genetics; Institut Pasteur; F-75015 Paris France
- CNRS; UMR3525 F-75015 Paris France
| |
Collapse
|
21
|
Baharoglu Z, Babosan A, Mazel D. Identification of genes involved in low aminoglycoside-induced SOS response in Vibrio cholerae: a role for transcription stalling and Mfd helicase. Nucleic Acids Res 2013; 42:2366-79. [PMID: 24319148 PMCID: PMC3936754 DOI: 10.1093/nar/gkt1259] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sub-inhibitory concentrations (sub-MIC) of antibiotics play a very important role in selection and development of resistances. Unlike Escherichia coli, Vibrio cholerae induces its SOS response in presence of sub-MIC aminoglycosides. A role for oxidized guanine residues was observed, but the mechanisms of this induction remained unclear. To select for V. cholerae mutants that do not induce low aminoglycoside-mediated SOS induction, we developed a genetic screen that renders induction of SOS lethal. We identified genes involved in this pathway using two strategies, inactivation by transposition and gene overexpression. Interestingly, we obtained mutants inactivated for the expression of proteins known to destabilize the RNA polymerase complex. Reconstruction of the corresponding mutants confirmed their specific involvement in induction of SOS by low aminoglycoside concentrations. We propose that DNA lesions formed on aminoglycoside treatment are repaired through the formation of single-stranded DNA intermediates, inducing SOS. Inactivation of functions that dislodge RNA polymerase leads to prolonged stalling on these lesions, which hampers SOS induction and repair and reduces viability under antibiotic stress. The importance of these mechanisms is illustrated by a reduction of aminoglycoside sub-MIC. Our results point to a central role for transcription blocking at DNA lesions in SOS induction, so far underestimated.
Collapse
Affiliation(s)
- Zeynep Baharoglu
- Département Génomes et Génétique, Unité Plasticité du Génome Bactérien, Institut Pasteur, 75015 Paris, France and Centre National de la Recherche Scientifique, CNRS, UMR3525 Paris, France
| | | | | |
Collapse
|
22
|
Wierstra I. The transcription factor FOXM1 (Forkhead box M1): proliferation-specific expression, transcription factor function, target genes, mouse models, and normal biological roles. Adv Cancer Res 2013; 118:97-398. [PMID: 23768511 DOI: 10.1016/b978-0-12-407173-5.00004-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor, which stimulates cell proliferation and exhibits a proliferation-specific expression pattern. Accordingly, both the expression and the transcriptional activity of FOXM1 are increased by proliferation signals, but decreased by antiproliferation signals, including the positive and negative regulation by protooncoproteins or tumor suppressors, respectively. FOXM1 stimulates cell cycle progression by promoting the entry into S-phase and M-phase. Moreover, FOXM1 is required for proper execution of mitosis. Accordingly, FOXM1 regulates the expression of genes, whose products control G1/S-transition, S-phase progression, G2/M-transition, and M-phase progression. Additionally, FOXM1 target genes encode proteins with functions in the execution of DNA replication and mitosis. FOXM1 is a transcriptional activator with a forkhead domain as DNA binding domain and with a very strong acidic transactivation domain. However, wild-type FOXM1 is (almost) inactive because the transactivation domain is repressed by three inhibitory domains. Inactive FOXM1 can be converted into a very potent transactivator by activating signals, which release the transactivation domain from its inhibition by the inhibitory domains. FOXM1 is essential for embryonic development and the foxm1 knockout is embryonically lethal. In adults, FOXM1 is important for tissue repair after injury. FOXM1 prevents premature senescence and interferes with contact inhibition. FOXM1 plays a role for maintenance of stem cell pluripotency and for self-renewal capacity of stem cells. The functions of FOXM1 in prevention of polyploidy and aneuploidy and in homologous recombination repair of DNA-double-strand breaks suggest an importance of FOXM1 for the maintenance of genomic stability and chromosomal integrity.
Collapse
|
23
|
Clark N, Wu X, Her C. MutS Homologues hMSH4 and hMSH5: Genetic Variations, Functions, and Implications in Human Diseases. Curr Genomics 2013; 14:81-90. [PMID: 24082819 PMCID: PMC3637681 DOI: 10.2174/1389202911314020002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/19/2013] [Accepted: 01/21/2013] [Indexed: 12/01/2022] Open
Abstract
The prominence of the human mismatch repair (MMR) pathway is clearly reflected by the causal link between MMR gene mutations and the occurrence of Lynch syndrome (or HNPCC). The MMR family of proteins also carries out a plethora of diverse cellular functions beyond its primary role in MMR and homologous recombination. In fact, members of the MMR family of proteins are being increasingly recognized as critical mediators between DNA damage repair and cell survival. Thus, a better functional understanding of MMR proteins will undoubtedly aid the development of strategies to effectively enhance apoptotic signaling in response to DNA damage induced by anti-cancer therapeutics. Among the five known human MutS homologs, hMSH4 and hMSH5 form a unique heterocomplex. However, the expression profiles of the two genes are not correlated in a number of cell types, suggesting that they may function independently as well. Consistent with this, these two proteins are promiscuous and thought to play distinct roles through interacting with different binding partners. Here, we describe the gene and protein structures of eukaryotic MSH4 and MSH5 with a particular emphasis on their human homologues, and we discuss recent findings of the roles of these two genes in DNA damage response and repair. Finally, we delineate the potential links of single nucleotide polymorphism (SNP) loci of these two genes with several human diseases.
Collapse
Affiliation(s)
- Nicole Clark
- STARS Program, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7520, USA ; School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7520, USA
| | | | | |
Collapse
|
24
|
Wu X, Xu Y, Feng K, Tompkins JD, Her C. MutS homologue hMSH5: recombinational DSB repair and non-synonymous polymorphic variants. PLoS One 2013; 8:e73284. [PMID: 24023853 PMCID: PMC3762724 DOI: 10.1371/journal.pone.0073284] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/18/2013] [Indexed: 01/25/2023] Open
Abstract
Double-strand breaks (DSBs) constitute the most deleterious form of DNA lesions that can lead to genome alterations and cell death, and the vast majority of DSBs arise pathologically in response to DNA damaging agents such as ionizing radiation (IR) and chemotherapeutic agents. Recent studies have implicated a role for the human MutS homologue hMSH5 in homologous recombination (HR)-mediated DSB repair and the DNA damage response. In the present study, we show that hMSH5 promotes HR-based DSB repair, and this property resides in the carboxyl-terminal portion of the protein. Our results demonstrate that DSB-triggered hMSH5 chromatin association peaks at the proximal regions of the DSB and decreases gradually with increased distance from the break. Furthermore, the DSB-triggered hMSH5 chromatin association is preceded by and relies on the assembly of hMRE11 and hRad51 at the proximal regions of the DSB. Lastly, the potential effects of hMSH5 non-synonymous variants (L85F, Y202C, V206F, R351G, L377F, and P786S) on HR and cell survival in response to DSB-inducing anticancer agents have been analyzed. These experiments show that the expression of hMSH5 variants elicits different survival responses to anticancer drugs cisplatin, bleomycin, doxorubicin and camptothecin. However, the effects of hMSH5 variants on survival responses to DSB-inducing agents are not directly correlated to their effects exerted on HR-mediated DSB repair, suggesting that the roles of hMSH5 variants in the processes of DNA damage response and repair are multifaceted.
Collapse
Affiliation(s)
- Xiling Wu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Yang Xu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Katey Feng
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Joshua D. Tompkins
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Chengtao Her
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| |
Collapse
|
25
|
Castro E, Olmos D, Garcia A, Cruz JJ, González-Sarmiento R. Role of XRCC3, XRCC1 and XPD single-nucleotide polymorphisms in survival outcomes following adjuvant chemotherapy in early stage breast cancer patients. Clin Transl Oncol 2013; 16:158-65. [DOI: 10.1007/s12094-013-1055-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 05/13/2013] [Indexed: 10/26/2022]
|
26
|
Wierstra I. FOXM1 (Forkhead box M1) in tumorigenesis: overexpression in human cancer, implication in tumorigenesis, oncogenic functions, tumor-suppressive properties, and target of anticancer therapy. Adv Cancer Res 2013; 119:191-419. [PMID: 23870513 DOI: 10.1016/b978-0-12-407190-2.00016-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor and is also intimately involved in tumorigenesis. FOXM1 stimulates cell proliferation and cell cycle progression by promoting the entry into S-phase and M-phase. Additionally, FOXM1 is required for proper execution of mitosis. In accordance with its role in stimulation of cell proliferation, FOXM1 exhibits a proliferation-specific expression pattern and its expression is regulated by proliferation and anti-proliferation signals as well as by proto-oncoproteins and tumor suppressors. Since these factors are often mutated, overexpressed, or lost in human cancer, the normal control of the foxm1 expression by them provides the basis for deregulated FOXM1 expression in tumors. Accordingly, FOXM1 is overexpressed in many types of human cancer. FOXM1 is intimately involved in tumorigenesis, because it contributes to oncogenic transformation and participates in tumor initiation, growth, and progression, including positive effects on angiogenesis, migration, invasion, epithelial-mesenchymal transition, metastasis, recruitment of tumor-associated macrophages, tumor-associated lung inflammation, self-renewal capacity of cancer cells, prevention of premature cellular senescence, and chemotherapeutic drug resistance. However, in the context of urethane-induced lung tumorigenesis, FOXM1 has an unexpected tumor suppressor role in endothelial cells because it limits pulmonary inflammation and canonical Wnt signaling in epithelial lung cells, thereby restricting carcinogenesis. Accordingly, FOXM1 plays a role in homologous recombination repair of DNA double-strand breaks and maintenance of genomic stability, that is, prevention of polyploidy and aneuploidy. The implication of FOXM1 in tumorigenesis makes it an attractive target for anticancer therapy, and several antitumor drugs have been reported to decrease FOXM1 expression.
Collapse
|
27
|
Abstract
DNA mismatch repair (MMR) corrects replication errors in newly synthesized DNA. It also has an antirecombination action on heteroduplexes that contain similar but not identical sequences. This review focuses on the genetics and development of MMR and not on the latest biochemical mechanisms. The main focus is on MMR in Escherichia coli, but examples from Streptococcuspneumoniae and Bacillussubtilis have also been included. In most organisms, only MutS (detects mismatches) and MutL (an endonuclease) and a single exonucleaseare present. How this system discriminates between newlysynthesized and parental DNA strands is not clear. In E. coli and its relatives, however, Dam methylation is an integral part of MMR and is the basis for strand discrimination. A dedicated site-specific endonuclease, MutH, is present, andMutL has no endonuclease activity; four exonucleases can participate in MMR. Although it might seem that the accumulated wealth of genetic and biochemical data has given us a detailed picture of the mechanism of MMR in E. coli, the existence of three competing models to explain the initiation phase indicates the complexity of the system. The mechanism of the antirecombination action of MMR is largely unknown, but only MutS and MutL appear to be necessary. A primary site of action appears to be on RecA, although subsequent steps of the recombination process can also be inhibited. In this review, the genetics of Very Short Patch (VSP) repair of T/G mismatches arising from deamination of 5-methylcytosineresidues is also discussed.
Collapse
|
28
|
Saleh EM, El-Awady RA, Anis N, El-Sharkawy N. Induction and repair of DNA double-strand breaks using constant-field gel electrophoresis and apoptosis as predictive markers for sensitivity of cancer cells to cisplatin. Biomed Pharmacother 2012; 66:554-62. [PMID: 22939736 DOI: 10.1016/j.biopha.2012.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 07/16/2012] [Accepted: 07/16/2012] [Indexed: 11/30/2022] Open
Abstract
This study was designed to evaluate some parameters that may play a role in the prediction of cancer cells sensitivity to cisplatin (CIS). Sensitivity, induction and repair of DNA double-strand breaks (DSB), cell cycle regulation and induction of apoptosis were measured in four cancer cell lines with different sensitivities to CIS. Using a sulphorhodamine-B assay, the cervical carcinoma cells (HeLa) were found to be the most sensitive to CIS followed by breast carcinoma cells (MCF-7) and liver carcinoma cells (HepG2). Colon carcinoma HCT116 cells were the most resistant. As measured by constant-field gel electrophoresis (CFGE), DSB induction, but not residual DSB exhibited a significant correlation with the sensitivity of cells to CIS. Flow cytometric DNA ploidy analysis revealed that 67% of HeLa cells and 10% of MCF-7 cells shift to sub-G1 phase after incubation with CIS. Additionally, CIS induced the arrest of MCF-7 cells in S-phase and the arrest of HepG2 and HCT116 cells in both S phase and G2/M phase. Determination of the Fas-L level and Caspase-9 activity indicated that CIS-induced apoptosis results from the mitochondrial (intrinsic) pathway. These results, if confirmed using clinical samples, indicate that the induction of DNA DSB as measured by CFGE and the induction of apoptosis should be considered, along with other predictive markers, in future clinical trials to develop predictive assays for platinum -based therapy.
Collapse
Affiliation(s)
- Ekram M Saleh
- Clinical Biochemistry and Molecular Biology unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| | | | | | | |
Collapse
|
29
|
Electron transfer-based combination therapy of cisplatin with tetramethyl-p-phenylenediamine for ovarian, cervical, and lung cancers. Proc Natl Acad Sci U S A 2012; 109:10175-80. [PMID: 22685209 DOI: 10.1073/pnas.1203451109] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The platinum-based chemotherapy is the standard treatment for several types of cancer. However, cancer cells often become refractory with time and most patients with serious cancers die of drug resistance. Recently, we have discovered a unique dissociative electron-transfer mechanism of action of cisplatin, the first and most widely used platinum-based anticancer drug. Here, we show that the combination of cisplatin with an exemplary biological electron donor, N,N,N',N'-tetramethyl-p-phenylenediamine (TMPD), may overcome the resistance of cancer cells to cisplatin. Our steady-state absorption and fluorescence spectroscopic measurements confirm the effective dissociative electron-transfer reaction between TMPD and cisplatin. More significantly, we found that the combination of 100 μM TMPD with cisplatin enhances double-strand breaks of plasmid DNA by a factor of approximately 3.5 and dramatically reduces the viability of cisplatin-sensitive human cervical (HeLa) cancer cells and highly cisplatin-resistant human ovarian (NIH:OVCAR-3) and lung (A549) cancer cells. Furthermore, this combination enhances apoptosis and DNA fragmentation by factors of 2-5 compared with cisplatin alone. These results demonstrate that this combination treatment not only results in a strong synergetic effect, but also makes resistant cancer cells sensitive to cisplatin. Because cisplatin is the cornerstone agent for the treatment of a variety of human cancers (including testicular, ovarian, cervical, bladder, head/neck, and lung cancers), our results show both the potential to improve platinum-based chemotherapy of various human cancers and the promise of femtomedicine as an emerging frontier in advancing cancer therapy.
Collapse
|
30
|
Xu T, Brown W, Marinus MG. Bleomycin sensitivity in Escherichia coli is medium-dependent. PLoS One 2012; 7:e33256. [PMID: 22438905 PMCID: PMC3305319 DOI: 10.1371/journal.pone.0033256] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 02/06/2012] [Indexed: 11/19/2022] Open
Abstract
Bleomycin (BLM) is a glycopeptide antibiotic and anti-tumor agent that targets primarily the furanose rings of DNA and in the presence of ferrous ions produces oxidative damage and DNA strand breaks. Escherichia coli cells growing in broth medium and exposed to low concentrations of BLM contain double-strand breaks and require homologous recombination to survive. To a lesser extent, the cells also require the abasic (AP) endonucleases associated with base excision repair, presumably to repair oxidative damage. As expected, there is strong induction of the SOS system in treated cells. In contrast, E. coli cells growing in glucose or glycerol minimal medium are resistant to the lethal action of BLM and do not require either homologous recombination functions or AP-endonucleases for survival. DNA ligase activity, however, is needed for cells growing in minimal medium to resist the lethal effects of BLM. There is weak SOS induction in such treated cells.
Collapse
Affiliation(s)
| | | | - Martin G. Marinus
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Tompkins JD, Wu X, Her C. MutS homologue hMSH5: role in cisplatin-induced DNA damage response. Mol Cancer 2012; 11:10. [PMID: 22401567 PMCID: PMC3325843 DOI: 10.1186/1476-4598-11-10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 03/08/2012] [Indexed: 11/10/2022] Open
Abstract
Background Cisplatin (cis-diamminedichloroplatinum (II), CDDP) and its analogues constitute an important class of anticancer drugs in the treatment of various malignancies; however, its effectiveness is frequently affected by mutations in genes involved in the repair and signaling of cisplatin-induced DNA damage. These observations necessitate a need for a better understanding of the molecular events governing cellular sensitivity to cisplatin. Results Here, we show that hMSH5 mediates sensitization to cisplatin-induced DNA damage in human cells. Our study indicates that hMSH5 undergoes cisplatin-elicited protein induction and tyrosine phosphorylation. Silencing of hMSH5 by RNAi or expression of hMSH5 phosphorylation-resistant mutant hMSH5Y742F elevates cisplatin-induced G2 arrest and renders cells susceptible to cisplatin toxicity at clinically relevant doses. In addition, our data show that cisplatin promotes hMSH5 chromatin association and hMSH5 deficiency increases cisplatin-triggered γ-H2AX foci. Consistent with a possible role for hMSH5 in recombinational repair of cisplatin-triggered double-strand breaks (DSBs), the formation of cisplatin-induced hMSH5 nuclear foci is hRad51-dependent. Conclusion Collectively, our current study has suggested a role for hMSH5 in the processing of cisplatin-induced DSBs, and silencing of hMSH5 may provide a new means to improve the therapeutic efficacy of cisplatin.
Collapse
Affiliation(s)
- Joshua D Tompkins
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Mail Drop 64-7520, Pullman, WA 99164, USA
| | | | | |
Collapse
|
32
|
Sharma A, Shukla A, Mishra M, Chowdhuri DK. Validation and application of Drosophila melanogaster as an in vivo model for the detection of double strand breaks by neutral Comet assay. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2011; 721:142-6. [DOI: 10.1016/j.mrgentox.2011.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 01/05/2011] [Accepted: 01/16/2011] [Indexed: 10/18/2022]
|
33
|
Danesi CC, Bellagamba BC, Dihl RR, Andrade HHRD, Cunha KS, Lehmann M. Evaluation of the genotoxicity of cisplatin, paclitaxel and 5-fluorouracil combined treatment in the Drosophila wing-spot test. Food Chem Toxicol 2010; 48:3120-4. [DOI: 10.1016/j.fct.2010.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 08/03/2010] [Accepted: 08/11/2010] [Indexed: 12/12/2022]
|
34
|
Becket E, Chen F, Tamae C, Miller JH. Determination of hypersensitivity to genotoxic agents among Escherichia coli single gene knockout mutants. DNA Repair (Amst) 2010; 9:949-57. [PMID: 20674514 PMCID: PMC2929269 DOI: 10.1016/j.dnarep.2010.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2010] [Revised: 05/19/2010] [Accepted: 06/17/2010] [Indexed: 10/19/2022]
Abstract
We have tested the KEIO collection of 3985 different viable single gene knockouts in Escherichia coli to identify genes whose loss increases sensitivity to one or more of six different chemotherapeutic agents and mutagens: Bleomycin (BLM), Cisplatin (CPT), ICR-191 (ICR), 5-azacytidine (5AZ), Zebularine (ZEB), and 5-bromo-2'-deoxyuridine (5BdU). We discovered a set of 156 strains that display a significant increase in sensitivity to at least one of the agents tested. Each genotoxic agent generates a distinct "sensitivity profile" that is characteristic of the agent. Comparison with an independent study of sensitivity profiles for an extensive set of antibiotics pinpoints those effects that are relatively specific for each agent. In some cases engineered double mutants have greatly increased effects. These results provide insight into the mechanism of action of each agent, and define targets for the design of co-drugs that can potentiate these agents. An example is the finding that mutants lacking one of several genes in the folate biosynthetic pathway are hypersensitive to ZEB, leading to a demonstration of synergy between trimethoprim and ZEB.
Collapse
Affiliation(s)
- Elinne Becket
- Department of Microbiology, Immunology, and Molecular Genetics, and The Molecular Biology Institute, University of California, and The David Geffen School of Medicine, Los Angeles, CA 90095
| | - Frank Chen
- Department of Microbiology, Immunology, and Molecular Genetics, and The Molecular Biology Institute, University of California, and The David Geffen School of Medicine, Los Angeles, CA 90095
| | - Cindy Tamae
- Department of Microbiology, Immunology, and Molecular Genetics, and The Molecular Biology Institute, University of California, and The David Geffen School of Medicine, Los Angeles, CA 90095
| | - Jeffrey H. Miller
- Department of Microbiology, Immunology, and Molecular Genetics, and The Molecular Biology Institute, University of California, and The David Geffen School of Medicine, Los Angeles, CA 90095
| |
Collapse
|
35
|
Kwok JMM, Peck B, Monteiro LJ, Schwenen HDC, Millour J, Coombes RC, Myatt SS, Lam EWF. FOXM1 confers acquired cisplatin resistance in breast cancer cells. Mol Cancer Res 2010; 8:24-34. [PMID: 20068070 DOI: 10.1158/1541-7786.mcr-09-0432] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The transcription factor Forkhead box M1 (FOXM1) is a key regulator of cell proliferation and is overexpressed in many forms of primary cancers, leading to uncontrolled cell division and genomic instability. To address the role of FOXM1 in chemoresistance, we generated a cisplatin-resistant breast cancer cell line (MCF-7-CIS(R)), which had an elevated level of FOXM1 protein and mRNA expression relative to the parental MCF-7 cells. A close correlation was observed between FOXM1 and the expression of its proposed downstream targets that are involved in DNA repair; breast cancer-associated gene 2 (BRCA2) and X-ray cross-complementing group 1 (XRCC1) were expressed at higher levels in the resistant cell lines compared with the sensitive MCF-7 cells. Moreover, cisplatin treatment induced DNA damage repair in MCF-7-CIS(R) and not in MCF-7 cells. Furthermore, the expression of a constitutively active FOXM1 (DeltaN-FOXM1) in MCF-7 cells alone was sufficient to confer cisplatin resistance. Crucially, the impairment of DNA damage repair pathways through the small interfering RNA knockdown inhibition of either FOXM1 or BRCA2/XRCC1 showed that only the silencing of FOXM1 could significantly reduce the rate of proliferation in response to cisplatin treatment in the resistant cells. This suggests that the targeting of FOXM1 is a viable strategy in circumventing acquired cisplatin resistance. Consistently, the FOXM1 inhibitor thiostrepton also showed efficacy in causing cell death and proliferative arrest in the cisplatin-resistant cells through the downregulation of FOXM1 expression. Taken together, we have identified a novel mechanism of acquired cisplatin resistance in breast cancer cells through the induction of FOXM1.
Collapse
Affiliation(s)
- Jimmy M-M Kwok
- Cancer Research-UK Laboratories, Department of Oncology, Medical Research Council Cyclotron Building, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Khan NUH, Pandya N, Kureshy RI, Abdi SHR, Agrawal S, Bajaj HC, Pandya J, Gupte A. Synthesis, characterization, DNA binding and cleavage studies of chiral Ru(II) salen complexes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2009; 74:113-119. [PMID: 19523873 DOI: 10.1016/j.saa.2009.05.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/14/2009] [Accepted: 05/14/2009] [Indexed: 05/27/2023]
Abstract
Interaction of chiral Ru(II) salen complexes (S)-1 and (R)-1 with Calf Thymus DNA (CT-DNA) was studied by absorption spectroscopy, competitive binding study, viscosity measurements, CD measurements, thermal denaturation study and cleavage studies by agarose gel electrophoresis. The DNA binding affinity of (S)-1 (6.25 x 10(3)M(-1)) was found to be greater than (R)-1 (3.0 x 10(3)M(-1)). The antimicrobial studies of these complexes on five different gram (+)/(-) bacteria and three different fungal organisms showed selective inhibition of the growth of gram (+) bacteria and were not affective against gram (-) and fungal organisms. Further, the (S)-1 enantiomer inhibited the growth of organisms to a greater extent as compared to (R)-1 enantiomer.
Collapse
Affiliation(s)
- Noor-ul H Khan
- Discipline of Inorganic Materials and Catalysis, Central Salt and Marine Chemicals Research Institute, Council of Scientific & Industrial Research, GB Marg, Bhavnagar 364002, Gujarat, India.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The DNA of Escherichia coli contains 19,120 6-methyladenines and 12,045 5-methylcytosines in addition to the four regular bases, and these are formed by the postreplicative action of three DNA methyltransferases. The majority of the methylated bases are formed by the Dam and Dcmmethyltransferases encoded by the dam (DNA adenine methyltransferase) and dcm (DNA cytosine methyltransferase) genes. Although not essential, Dam methylation is important for strand discrimination during repair of replication errors, controlling the frequency of initiation of chromosome replication at oriC, and regulation of transcription initiation at promoters containing GATC sequences. In contrast, there is no known function for Dcm methylation, although Dcm recognition sites constitute sequence motifs for Very Short Patch repair of T/G base mismatches. In certain bacteria (e.g., Vibrio cholera and Caulobactercrescentus) adenine methylation is essential, and in C.crescentus it is important for temporal gene expression which, in turn, is required for coordination of chromosome initiation, replication, and division. In practical terms, Dam and Dcm methylation can inhibit restriction enzyme cleavage,decrease transformation frequency in certain bacteria,and decrease the stability of short direct repeats andare necessary for site-directed mutagenesis and to probe eukaryotic structure and function.
Collapse
|
38
|
Poteete AR. Expansion of a chromosomal repeat in Escherichia coli: roles of replication, repair, and recombination functions. BMC Mol Biol 2009; 10:14. [PMID: 19236706 PMCID: PMC2656507 DOI: 10.1186/1471-2199-10-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 02/23/2009] [Indexed: 11/21/2022] Open
Abstract
Background Previous studies of gene amplification in Escherichia coli have suggested that it occurs in two steps: duplication and expansion. Expansion is thought to result from homologous recombination between the repeated segments created by duplication. To explore the mechanism of expansion, a 7 kbp duplication in the chromosome containing a leaky mutant version of the lac operon was constructed, and its expansion into an amplified array was studied. Results Under selection for lac function, colonies bearing multiple copies of the mutant lac operon appeared at a constant rate of approximately 4 to 5 per million cells plated per day, on days two through seven after plating. Expansion was not seen in a recA strain; null mutations in recBCD and ruvC reduced the rate 100- and 10-fold, respectively; a ruvC recG double mutant reduced the rate 1000-fold. Expansion occurred at an increased rate in cells lacking dam, polA, rnhA, or uvrD functions. Null mutations of various other cellular recombination, repair, and stress response genes had little effect upon expansion. The red recombination genes of phage lambda could substitute for recBCD in mediating expansion. In the red-substituted cells, expansion was only partially dependent upon recA function. Conclusion These observations are consistent with the idea that the expansion step of gene amplification is closely related, mechanistically, to interchromosomal homologous recombination events. They additionally provide support for recently described models of RecA-independent Red-mediated recombination at replication forks.
Collapse
Affiliation(s)
- Anthony R Poteete
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
39
|
Lin CT, Lai HC, Lee HY, Lin WH, Chang CC, Chu TY, Lin YW, Lee KD, Yu MH. Valproic acid resensitizes cisplatin-resistant ovarian cancer cells. Cancer Sci 2008; 99:1218-26. [PMID: 18429963 PMCID: PMC11160082 DOI: 10.1111/j.1349-7006.2008.00793.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Revised: 01/17/2008] [Accepted: 01/31/2008] [Indexed: 11/28/2022] Open
Abstract
Although certain inhibitors of histone deacetylases have been shown to induce cytotoxicity alone or in combination with chemotherapeutic agents in cancer cells, the molecular mechanism is not clear. The goal of the present study was to determine whether the antiseizure drug valproic acid (2-propylpentanoic acid; VPA), which is also able to inhibit histone deacetylase, exhibits synergistic cytotoxicity with cisplatin, and the possible pathways for this. Our results clearly show that VPA not only exhibits synergistic cytotoxicity with cisplatin in all of the ovarian carcinoma cells tested, but also can resensitize the cells that have acquired resistance to cisplatin. Consistent with the increased cytotoxicity, cotreatment with VPA was shown to upregulate the cisplatin-mediated DNA damage revealed by phosphorylation of ataxia telangiectasia mutation and histone H2AX. Reactive oxygen species accumulation and tumor suppressor phosphatase and tensin homolog (PTEN) overexpression, which could contribute to the enhanced cytotoxicity, were also observed to be upregulated by VPA. Because PTEN knockdown by small interference RNA or antioxidant treatment can reduce cisplatin-mediated cytotoxicity, it is suggested that upregulation of PTEN and reactive oxygen species by VPA contributes to the enhancement of cisplatin-mediated cytotoxicity. These results with resensitization of cisplatin-resistant cells particularly may provide benefits in the treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Ching-Tai Lin
- Department of Medical Research, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Activities of DNA-PK and Ku86, but not Ku70, may predict sensitivity to cisplatin in human gliomas. J Neurooncol 2008; 89:27-35. [PMID: 18415044 DOI: 10.1007/s11060-008-9592-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 03/28/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE This study was designed to investigate the relationship between activities of DNA-dependent protein kinase (DNA-PK), its subunits Ku86/Ku70, and sensitivities to cisplatin in human glioma samples. METHODS Thirty-six glioma samples from patients without prior treatment before neurosurgery were included in this study. The sensitivities to cisplatin as indicated by IC(50) (the inhibitory concentration leading to 50% cell death) were assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenytetrazolium (MTT) assay; activities of DNA-PK and Ku70/Ku86 were analyzed by SigmaTECT DNA-Dependent Protein Kinase Assay System and Ku70/Ku86 DNA Repair Kit, respectively. RESULTS Sensitivities to cisplatin correlated with the activities of DNA-PK/Ku86, but not with the Ku70 or other clinical parameters such as age, sex of the patients, pathological gradings of the tumors, or tumor size. The levels of DNA-PK activities also associated with pathological grading and Ku86, but not with other clinical parameters. The tumors of the patients who failed to respond to cisplatin-based chemotherapy tended to display higher activity levels of DNA-PK and Ku86. Furthermore, platinum-based chemotherapy did not result in significant changes of DNA-PK/Ku activities in four matched samples before and after chemotherapy. CONCLUSION Pretreatment determination of DNA-PK/Ku86 activities might be helpful in identifying patients who will actually benefit from platinum-based treatment.
Collapse
|
41
|
Murphy KC, Ritchie JM, Waldor MK, Løbner-Olesen A, Marinus MG. Dam methyltransferase is required for stable lysogeny of the Shiga toxin (Stx2)-encoding bacteriophage 933W of enterohemorrhagic Escherichia coli O157:H7. J Bacteriol 2008; 190:438-41. [PMID: 17981979 PMCID: PMC2223730 DOI: 10.1128/jb.01373-07] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Accepted: 10/18/2007] [Indexed: 11/20/2022] Open
Abstract
Shiga toxin 2 (Stx2), one of the principal virulence factors of enterohemorrhagic Escherichia coli, is encoded by 933W, a lambda-like prophage. 933W prophage induction contributes to Stx2 production, and here, we provide evidence that Dam methyltransferase is essential for maintenance of 933W lysogeny. Our findings are consistent with the idea that the 933W prophage has a relatively low threshold for induction, which may promote Stx2 production during infection.
Collapse
Affiliation(s)
- Kenan C Murphy
- University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
42
|
Nowosielska A, Marinus MG. DNA mismatch repair-induced double-strand breaks. DNA Repair (Amst) 2008; 7:48-56. [PMID: 17827074 PMCID: PMC2175267 DOI: 10.1016/j.dnarep.2007.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 05/21/2007] [Accepted: 07/18/2007] [Indexed: 11/26/2022]
Abstract
Escherichia coli dam mutants are sensitized to the cytotoxic action of base analogs, cisplatin and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), while their mismatch repair (MMR)-deficient derivatives are tolerant to these agents. We showed previously, using pulse field gel electrophoresis (PFGE), that MMR-mediated double-strand breaks (DSBs) are produced by cisplatin in dam recB(Ts) cells at the non-permissive temperature. We demonstrate here that the majority of these DSBs require DNA replication for their formation, consistent with a model in which replication forks collapse at nicks or gaps formed during MMR. DSBs were also detected in dam recB(Ts) ada ogt cells exposed to MNNG in a dose- and MMR-dependent manner. In contrast to cisplatin, the formation of these DSBs was not affected by DNA replication and it is proposed that two separate mechanisms result in DSB formation. Replication-independent DSBs arise from overlapping base excision and MMR repair tracts on complementary strands and constitute the majority of detectable DSBs in dam recB(Ts) ada ogt cells exposed to MNNG. Replication-dependent DSBs result from replication fork collapse at O(6)-methylguanine (O(6)-meG) base pairs undergoing MMR futile cycling and are more likely to contribute to cytotoxicity. This model is consistent with the observation that fast-growing dam recB(Ts) ada ogt cells, which have more chromosome replication origins, are more sensitive to the cytotoxic effect of MNNG than the same cells growing slowly.
Collapse
Affiliation(s)
- Anetta Nowosielska
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | | |
Collapse
|
43
|
Centore RC, Lestini R, Sandler SJ. XthA (Exonuclease III) regulates loading of RecA onto DNA substrates in log phase Escherichia coli cells. Mol Microbiol 2007; 67:88-101. [DOI: 10.1111/j.1365-2958.2007.06026.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Jacquelín DK, Martina MA, Argaraña CE, Barra JL. Plasmid expression of mutS, -L and/or -H gene in Escherichia coli dam cells results in strains that display reduced mutation frequency. Mutat Res 2007; 637:197-204. [PMID: 17727900 DOI: 10.1016/j.mrfmmm.2007.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2007] [Revised: 06/29/2007] [Accepted: 07/17/2007] [Indexed: 11/20/2022]
Abstract
Escherichia colidam cells have an active but non-directed mismatch repair system; therefore, assembly of MutSLH complex at a mismatched base pair can result in MutH-mediated cleavage of GATC sites in both DNA strands. Unpaired double-strand breaks on a fraction of the replication errors occurring in dam cells presumably cause cell death, selectively eliminating these putative mutants from the population. We show that E. colidam cells transformed with plasmids containing either the mutS, mutL or mutH gene display a mutation frequency three to eight times lower than that of the parental dam strain, due to increased mismatch-stimulated cell killing. Transformed strains are also more susceptible to killing by the base analogue 2-aminopurine. However, dam and dam transformed cells have similar duplication time, proportion of live/dead cells and morphology.
Collapse
Affiliation(s)
- Daniela K Jacquelín
- Centro de Investigaciones en Química Biológica de Córdoba, Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | | | | | | |
Collapse
|
45
|
Lu QB, Kalantari S, Wang CR. Electron transfer reaction mechanism of cisplatin with DNA at the molecular level. Mol Pharm 2007; 4:624-8. [PMID: 17559229 DOI: 10.1021/mp070040a] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The cytotoxicity of cisplatin as a very effective chemotherapeutic anticancer drug is known to arise from its capacity to damage DNA. However, the mechanism of action of cisplatin at the molecular level, in particular, the reaction dynamics of cisplatin with DNA, remains elusive, and the reason why cisplatin binds to the guanine bases rather than to S-donor ligands available in cells remains a mystery. Using time-resolved femtosecond laser spectroscopy, for the first time, we reveal the high reactivity of cisplatin with electrons and its preferential electron-transfer reaction with the DNA guanine base. The results not only provide a molecular mechanistic understanding of cisplatin in cancer therapy but may have far-reaching significance for understanding the role of polar molecules such as NH3 and NH2 in biological reactions.
Collapse
Affiliation(s)
- Q-B Lu
- Department of Physics, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada.
| | | | | |
Collapse
|
46
|
Lu QB. Molecular reaction mechanisms of combination treatments of low-dose cisplatin with radiotherapy and photodynamic therapy. J Med Chem 2007; 50:2601-4. [PMID: 17474733 DOI: 10.1021/jm061416b] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Combination of low-dose cisplatin with radiotherapy or photodynamic therapy (PDT) is a novel cancer treatment. Using time-resolved femtosecond laser spectroscopy, we reveal the molecular mechanisms of the combinations of cisplatin with radiotherapy and PDT using indocyanine green (ICG) excited at 800 nm. DNA damage measurements confirm that electron-transfer reactions of cisplatin with electrons generated in ionizing radiation and with the ICG singlet excited state in PDT are responsible for the cytotoxic enhancements.
Collapse
Affiliation(s)
- Q-B Lu
- Departments of Physics and Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada.
| |
Collapse
|
47
|
Prieto AI, Ramos-Morales F, Casadesús J. Repair of DNA damage induced by bile salts in Salmonella enterica. Genetics 2006; 174:575-84. [PMID: 16888329 PMCID: PMC1602091 DOI: 10.1534/genetics.106.060889] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exposure of Salmonella enterica to sodium cholate, sodium deoxycholate, sodium chenodeoxycholate, sodium glycocholate, sodium taurocholate, or sodium glycochenodeoxycholate induces the SOS response, indicating that the DNA-damaging activity of bile resides in bile salts. Bile increases the frequency of GC --> AT transitions and induces the expression of genes belonging to the OxyR and SoxRS regulons, suggesting that bile salts may cause oxidative DNA damage. S. enterica mutants lacking both exonuclease III (XthA) and endonuclease IV (Nfo) are bile sensitive, indicating that S. enterica requires base excision repair (BER) to overcome DNA damage caused by bile salts. Bile resistance also requires DinB polymerase, suggesting the need of SOS-associated translesion DNA synthesis. Certain recombination functions are also required for bile resistance, and a key factor is the RecBCD enzyme. The extreme bile sensitivity of RecB-, RecC-, and RecA- RecD- mutants provides evidence that bile-induced damage may impair DNA replication.
Collapse
Affiliation(s)
- Ana I Prieto
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville 41080, Spain
| | | | | |
Collapse
|
48
|
Sutera VA, Lovett ST. The role of replication initiation control in promoting survival of replication fork damage. Mol Microbiol 2006; 60:229-39. [PMID: 16556234 DOI: 10.1111/j.1365-2958.2006.05093.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dam methylase mutants were recovered in a screen for mutants sensitive to UV irradiation or mild inhibition of replication elongation. Dam's role in tolerance of DNA damage is to provide binding sites for SeqA, because seqA mutants showed similar sensitivity that was genetically epistatic to dam. The sensitivity of seqA mutants to UV irradiation and to the replication inhibitors hydroxyurea (HU) and azidothymidine (AZT) was suppressed by alleles of dnaA that reduce the efficiency of replication initiation. These results suggest that for survival of replication fork damage, SeqA's repression of replication initiation is more important than its effects on nucleoid organization. Convergence of forks upon DNA damage is a likely explanation for seqA mutant sensitivity, because its poor survival of UV was suppressed by reducing secondary initiation through minimal medium growth. Surprisingly, growth in minimal medium reduced the ability of seqA+ strains to form colonies in the presence of low levels of AZT. Double dnaA seqA mutants exhibited plating efficiencies much superior to wild-type strains during chronic low-level AZT exposure in minimal medium. This suggests that mild inhibition of replication fork progression may actively restrain initiation such that seqA+ strains fail to recover initiation capacity after sustained conditions of replication arrest.
Collapse
Affiliation(s)
- Vincent A Sutera
- Department of Biology and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454-9110, USA
| | | |
Collapse
|
49
|
Boonsombat R, Yeh SP, Milne A, Sandler SJ. A novel dnaC mutation that suppresses priB rep mutant phenotypes in Escherichia coli K-12. Mol Microbiol 2006; 60:973-83. [PMID: 16677308 DOI: 10.1111/j.1365-2958.2006.05147.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The loading of a replisome in prokaryotic and eukaryotic cells at an origin of DNA replication and during replication restart is a highly ordered and regulated process. During replication restart in Escherichia coli, the PriA, PriB, PriC, DnaT and Rep proteins form multiple pathways that bind to repaired replication forks. These complexes are then recognized by DnaC as sites to load DnaB, the replicative helicase. Several dnaC mutations have been isolated that suppress phenotypes of some replication restart mutants. A new dnaC mutation (dnaC824) is reported here that efficiently suppresses priB rep mutant phenotypes. Furthermore, it is shown that dnaC824 will suppress phenotypes of priB priA300, rep priA300 and priB priC strains. Unlike other dnaC suppressors, it can only weakly suppress the absence of priA. Others have reported a different type of dnaC mutation, dnaC1331, is able to mimic priB mutant phenotypes. This is supported herein by showing that like dnaC1331, a priB mutation is synthetically lethal with a dam mutation and this can be rescued by a mutH mutation. Furthermore, priB dam lethality can also be suppressed by dnaC824. Like a priB mutation, a dnaC1331 mutation causes a priA2::kan-like phenotype when combined with priA300. Lastly, we show that dnaC824 is dominant to wild type and that dnaC1331 is recessive to wild type. Several models are discussed for the action of these mutant dnaC proteins in replication restart.
Collapse
Affiliation(s)
- Ruethairat Boonsombat
- Department of Microbiology, Morrill Science Center IV N203, University of Massachusetts at Amherst, Amherst, MA 01003, USA
| | | | | | | |
Collapse
|
50
|
Calmann MA, Marinus M. Differential effects of cisplatin and MNNG on dna mutants of Escherichia coli. Mutat Res 2005; 578:406-16. [PMID: 16144703 PMCID: PMC2927670 DOI: 10.1016/j.mrfmmm.2005.06.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Revised: 06/09/2005] [Accepted: 06/09/2005] [Indexed: 02/08/2023]
Abstract
DNA mismatch repair (MMR) in mammalian cells or Escherichia coli dam mutants increases the cytotoxic effects of cisplatin and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). We found that, unlike wildtype, the dnaE486 (alpha catalytic subunit of DNA polymerase III holoenzyme) mutant, and a DnaX (clamp loader subunits) over-producer, are sensitive to cisplatin but resistant to MNNG at the permissive temperature for growth. Survival of dam-13 dnaN159 (beta sliding clamp) bacteria to cisplatin was significantly less than dam cells, suggesting decreased MMR, which may be due to reduced MutS-beta clamp interaction. We also found an elevated spontaneous mutant frequency to rifampicin resistance in dnaE486 (10-fold), dnaN159 (35-fold) and dnaX36 (10-fold) strains. The mutation spectrum in the dnaN159 strain was consistent with increased SOS induction and not indicative of MMR deficiency.
Collapse
Affiliation(s)
- Melissa A. Calmann
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester MA 01655
| | - M.G. Marinus
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester MA 01655
| |
Collapse
|