1
|
Uthumange SS, Liew AJH, Chee XW, Yeong KY. Ringing medicinal chemistry: The importance of 3-membered rings in drug discovery. Bioorg Med Chem 2024; 116:117980. [PMID: 39536361 DOI: 10.1016/j.bmc.2024.117980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Scaffold-based drug design has become increasingly prominent in the pharmaceutical field due to the systematic and effective approach through which it facilitates the development of novel drugs. The identification of key scaffolds provides medicinal chemists with a fundamental framework for subsequent research. With mounting evidence suggesting that increased aromaticity could impede the chances of developmental success for oral drug candidates, there is an imperative need for a more thorough exploration of alternative ring systems to mitigate attrition risks. The unique characteristics exhibited by three-membered rings have led to their application in medicinal chemistry. This review explores the use of cyclopropane-, aziridine-, thiirane-, and epoxide-containing compounds in drug discovery, focusing on their roles in approved medicines and drug candidates. Specifically, the importance of the three-membered ring systems in rending biological activity for each drug molecule was highlighted. The undeniable therapeutic value and intriguing features presented by these compounds suggest significant pharmacological potential, providing justification for their incorporation into the design of novel drug candidates.
Collapse
Affiliation(s)
- Sahani Sandalima Uthumange
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor, Malaysia
| | - Angie Jun Hui Liew
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor, Malaysia
| | - Xavier Wezen Chee
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Keng Yoon Yeong
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor, Malaysia.
| |
Collapse
|
2
|
Gach-Janczak K, Drogosz-Stachowicz J, Janecka A, Wtorek K, Mirowski M. Historical Perspective and Current Trends in Anticancer Drug Development. Cancers (Basel) 2024; 16:1878. [PMID: 38791957 PMCID: PMC11120596 DOI: 10.3390/cancers16101878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer is considered one of the leading causes of death in the 21st century. The intensive search for new anticancer drugs has been actively pursued by chemists and pharmacologists for decades, focusing either on the isolation of compounds with cytotoxic properties from plants or on screening thousands of synthetic molecules. Compounds that could potentially become candidates for new anticancer drugs must have the ability to inhibit proliferation and/or induce apoptosis in cancer cells without causing too much damage to normal cells. Some anticancer compounds were discovered by accident, others as a result of long-term research. In this review, we have presented a brief history of the development of the most important groups of anticancer drugs, pointing to the fact that they all have many side effects.
Collapse
Affiliation(s)
- Katarzyna Gach-Janczak
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.J.); (K.W.)
| | | | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.J.); (K.W.)
| | - Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.J.); (K.W.)
| | - Marek Mirowski
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
3
|
Kamat A, Tran NT, Sharda M, Sontakke N, Le TBK, Badrinarayanan A. Widespread prevalence of a methylation-dependent switch to activate an essential DNA damage response in bacteria. PLoS Biol 2024; 22:e3002540. [PMID: 38466718 PMCID: PMC10957082 DOI: 10.1371/journal.pbio.3002540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/21/2024] [Accepted: 02/06/2024] [Indexed: 03/13/2024] Open
Abstract
DNA methylation plays central roles in diverse cellular processes, ranging from error-correction during replication to regulation of bacterial defense mechanisms. Nevertheless, certain aberrant methylation modifications can have lethal consequences. The mechanisms by which bacteria detect and respond to such damage remain incompletely understood. Here, we discover a highly conserved but previously uncharacterized transcription factor (Cada2), which orchestrates a methylation-dependent adaptive response in Caulobacter. This response operates independently of the SOS response, governs the expression of genes crucial for direct repair, and is essential for surviving methylation-induced damage. Our molecular investigation of Cada2 reveals a cysteine methylation-dependent posttranslational modification (PTM) and mode of action distinct from its Escherichia coli counterpart, a trait conserved across all bacteria harboring a Cada2-like homolog instead. Extending across the bacterial kingdom, our findings support the notion of divergence and coevolution of adaptive response transcription factors and their corresponding sequence-specific DNA motifs. Despite this diversity, the ubiquitous prevalence of adaptive response regulators underscores the significance of a transcriptional switch, mediated by methylation PTM, in driving a specific and essential bacterial DNA damage response.
Collapse
Affiliation(s)
- Aditya Kamat
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Ngat T. Tran
- John Innes Centre, Department of Molecular Microbiology, Colney Lane, Norwich, United Kingdom
| | - Mohak Sharda
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Neha Sontakke
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Tung B. K. Le
- John Innes Centre, Department of Molecular Microbiology, Colney Lane, Norwich, United Kingdom
| | | |
Collapse
|
4
|
Gessner S, Martin ZAM, Reiche MA, Santos JA, Dinkele R, Ramudzuli A, Dhar N, de Wet TJ, Anoosheh S, Lang DM, Aaron J, Chew TL, Herrmann J, Müller R, McKinney JD, Woodgate R, Mizrahi V, Venclovas Č, Lamers MH, Warner DF. Investigating the composition and recruitment of the mycobacterial ImuA'-ImuB-DnaE2 mutasome. eLife 2023; 12:e75628. [PMID: 37530405 PMCID: PMC10421592 DOI: 10.7554/elife.75628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 08/01/2023] [Indexed: 08/03/2023] Open
Abstract
A DNA damage-inducible mutagenic gene cassette has been implicated in the emergence of drug resistance in Mycobacterium tuberculosis during anti-tuberculosis (TB) chemotherapy. However, the molecular composition and operation of the encoded 'mycobacterial mutasome' - minimally comprising DnaE2 polymerase and ImuA' and ImuB accessory proteins - remain elusive. Following exposure of mycobacteria to DNA damaging agents, we observe that DnaE2 and ImuB co-localize with the DNA polymerase III β subunit (β clamp) in distinct intracellular foci. Notably, genetic inactivation of the mutasome in an imuBAAAAGG mutant containing a disrupted β clamp-binding motif abolishes ImuB-β clamp focus formation, a phenotype recapitulated pharmacologically by treating bacilli with griselimycin and in biochemical assays in which this β clamp-binding antibiotic collapses pre-formed ImuB-β clamp complexes. These observations establish the essentiality of the ImuB-β clamp interaction for mutagenic DNA repair in mycobacteria, identifying the mutasome as target for adjunctive therapeutics designed to protect anti-TB drugs against emerging resistance.
Collapse
Affiliation(s)
- Sophia Gessner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
| | - Zela Alexandria-Mae Martin
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
- Laboratory of Microbiology and Microsystems, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL)LausanneSwitzerland
| | - Michael A Reiche
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
- Advanced Imaging Center, Howard Hughes Medical InstituteAshburnUnited States
| | - Joana A Santos
- Department of Cell and Chemical Biology, Leiden University Medical CenterLeidenNetherlands
| | - Ryan Dinkele
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
| | - Atondaho Ramudzuli
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
| | - Neeraj Dhar
- Laboratory of Microbiology and Microsystems, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL)LausanneSwitzerland
| | - Timothy J de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
- Department of Integrative Biomedical Sciences, University of Cape TownCape TownSouth Africa
| | - Saber Anoosheh
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
| | - Dirk M Lang
- Confocal and Light Microscope Imaging Facility, Department of Human Biology, University of Cape TownCape TownSouth Africa
| | - Jesse Aaron
- Advanced Imaging Center, Howard Hughes Medical InstituteAshburnUnited States
| | - Teng-Leong Chew
- Advanced Imaging Center, Howard Hughes Medical InstituteAshburnUnited States
| | - Jennifer Herrmann
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
- German Centre for Infection Research (DZIF), Partner Site Hannover-BraunschweigBraunschweigGermany
| | - Rolf Müller
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
- German Centre for Infection Research (DZIF), Partner Site Hannover-BraunschweigBraunschweigGermany
| | - John D McKinney
- Laboratory of Microbiology and Microsystems, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL)LausanneSwitzerland
| | - Roger Woodgate
- Laboratory of Genomic Integrity, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaUnited States
| | - Valerie Mizrahi
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape TownCape TownSouth Africa
| | | | - Meindert H Lamers
- Department of Cell and Chemical Biology, Leiden University Medical CenterLeidenNetherlands
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, University of Cape TownCape TownSouth Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape TownCape TownSouth Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape TownCape TownSouth Africa
| |
Collapse
|
5
|
Joseph AM, Nahar K, Daw S, Hasan MM, Lo R, Le TBK, Rahman KM, Badrinarayanan A. Mechanistic insight into the repair of C8-linked pyrrolobenzodiazepine monomer-mediated DNA damage. RSC Med Chem 2022; 13:1621-1633. [PMID: 36561066 PMCID: PMC9749960 DOI: 10.1039/d2md00194b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
Pyrrolobenzodiazepines (PBDs) are naturally occurring DNA binding compounds that possess anti-tumor and anti-bacterial activity. Chemical modifications of PBDs can result in improved DNA binding, sequence specificity and enhanced efficacy. More recently, synthetic PBD monomers have shown promise as payloads for antibody drug conjugates and anti-bacterial agents. The precise mechanism of action of these PBD monomers and their role in causing DNA damage remains to be elucidated. Here we characterized the damage-inducing potential of two C8-linked PBD bi-aryl monomers in Caulobacter crescentus and investigated the strategies employed by cells to repair the same. We show that these compounds cause DNA damage and efficiently kill bacteria, in a manner comparable to the extensively used DNA cross-linking agent mitomycin-C (MMC). However, in stark contrast to MMC which employs a mutagenic lesion tolerance pathway, we implicate essential functions for error-free mechanisms in repairing PBD monomer-mediated damage. We find that survival is severely compromised in cells lacking nucleotide excision repair and to a lesser extent, in cells with impaired recombination-based repair. Loss of nucleotide excision repair leads to significant increase in double-strand breaks, underscoring the critical role of this pathway in mediating repair of PBD-induced DNA lesions. Together, our study provides comprehensive insights into how mono-alkylating DNA-targeting therapeutic compounds like PBD monomers challenge cell growth, and identifies the specific mechanisms employed by the cell to counter the same.
Collapse
Affiliation(s)
- Asha Mary Joseph
- National Centre for Biological Sciences (Tata Institute of Fundamental Research) Bangalore India
| | - Kazi Nahar
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London Franklin-Wilkins Building, 150 Stamford Street London SE1 9NH UK
| | - Saheli Daw
- National Centre for Biological Sciences (Tata Institute of Fundamental Research) Bangalore India
| | - Md Mahbub Hasan
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London Franklin-Wilkins Building, 150 Stamford Street London SE1 9NH UK
| | - Rebecca Lo
- John Innes Centre, Department of Molecular Microbiology Colney Lane Norwich NR4 7UH UK
| | - Tung B K Le
- John Innes Centre, Department of Molecular Microbiology Colney Lane Norwich NR4 7UH UK
| | - Khondaker Miraz Rahman
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London Franklin-Wilkins Building, 150 Stamford Street London SE1 9NH UK
| | - Anjana Badrinarayanan
- National Centre for Biological Sciences (Tata Institute of Fundamental Research) Bangalore India
| |
Collapse
|
6
|
Efthimiou I, Vlastos D, Triantafyllidis V, Eleftherianos A, Antonopoulou M. Investigation of the Genotoxicological Profile of Aqueous Betula pendula Extracts. PLANTS (BASEL, SWITZERLAND) 2022; 11:2673. [PMID: 36297697 PMCID: PMC9611029 DOI: 10.3390/plants11202673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Betula pendula belongs to the Betulaceae family and is most common in the northern hemisphere. Various birch species have exhibited antimicrobial, antioxidant and anticancer properties. In the present study, we investigated the genotoxic and cytotoxic activity as well as the antigenotoxic potential against the mutagenic agent mitomycin-C (MMC) of two commercial products, i.e., a Betula pendula aqueous leaf extract product (BE) and a Betula pendula product containing aqueous extract of birch leaves at a percentage of 94% and lemon juice at a percentage of 6% (BP) using the cytokinesis block micronucleus (CBMN) assay. The most prevalent compounds and elements of BE and BP were identified using UHPLC-MS and ICP-MS/MS, respectively. All mixtures of BE with MMC demonstrated a decrease in the MN frequencies, with the lowest and highest concentrations inducing a statistically significant antigenotoxic activity. BP lacked genotoxic potential, while it was cytotoxic in all concentrations. Its mixtures with MMC demonstrated statistically significant antigenotoxic activity only at the lowest concentration. UHPLC-MS and ICP-MS/MS showed the presence of various elements and phytochemicals. Our results reveal antigenotoxic and cytotoxic potential of both BE and BP, while the variations observed could indicate the importance of the interactions among different natural products and/or their compounds.
Collapse
Affiliation(s)
- Ioanna Efthimiou
- Department of Sustainable Agriculture, University of Patras, GR-30100 Agrinio, Greece
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research (HCMR), GR-19013 Anavyssos, Greece
| | - Dimitris Vlastos
- Department of Biology, Section of Genetics Cell Biology and Development, University of Patras, GR-26500 Patras, Greece
| | | | - Antonios Eleftherianos
- Akrokeramos Sewerage Laboratory, Athens Water Supply and Sewerage Company (EYDAP SA), GR-18755 Keratsini, Greece
| | - Maria Antonopoulou
- Department of Sustainable Agriculture, University of Patras, GR-30100 Agrinio, Greece
| |
Collapse
|
7
|
Moon OJ, Yoon CJ, Lee BR, Lee J. An Optimally Fabricated Platform Guides Cancer-Specific Activation of Chemotherapeutic Drugs and Toxicity-free Cancer Treatment. Adv Healthc Mater 2022; 11:e2200765. [PMID: 35670274 DOI: 10.1002/adhm.202200765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/12/2022] [Indexed: 11/08/2022]
Abstract
Cancer chemotherapeutic drugs such as doxorubicin, mitomycin C, and gemcitabine, which are mostly small synthetic molecules, are still clinically useful for cancer treatment. However, despite considerable therapeutic efficacy, severe toxicity-associated problems, which are mainly caused by the non-specific mode of action such as chromosomal DNA damage and interference in the DNA replication even in normal cells, remain unresolved and a major challenge for safer and thus more widespread adoption of chemotherapy. Here we developed an innovative platform through beneficially integrating core peptide units into highly-ordered, stable, and flexibly guest-adaptable structure of apoferritin, which simultaneously fulfills high-capacity loading of chemotherapeutic drugs compared with the case of FDA-approved antibody-drug conjugates, efficient drug targeting to cancer cells, and cancer cell-specific drug release and activation. This approach dramatically reduced drug toxicity to normal cells, significantly enhanced efficacy in in vivo cancer treatment without toxicity to normal organs of mice, and thus is expected to open up a novel clinical route to break through the limits of current cancer chemotherapy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ok Jeong Moon
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| | - Chul Joo Yoon
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| | - Bo-Ram Lee
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| | - Jeewon Lee
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| |
Collapse
|
8
|
Cheng SY, Delgado-Cruzata L, Clement CC, Zacarias O, Concheiro-Guisan M, Towler N, Snyder T, Zheng M, Almodovar N, Gonzalez C, Romaine M, Sapse AM, Champeil E. Cytotoxicity, crosslinking and biological activity of three mitomycins. Bioorg Chem 2022; 123:105744. [PMID: 35349830 PMCID: PMC9050950 DOI: 10.1016/j.bioorg.2022.105744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/17/2022] [Accepted: 03/13/2022] [Indexed: 11/30/2022]
Abstract
While interstrand crosslinks (ICLs) have been considered as one type of DNA damage in the past, there is mounting evidence suggesting that these highly cytotoxic lesions are processed differently by the cellular machinery depending upon the ICL structure. In this study, we examined the crosslinking ability of three mitomycins, the structure of the ICLs they produce and the cytotoxicity of the drugs toward three different cell lines. The drugs are: mitomycin C (1), decarbamoylmitomycin C (2), and a mitomycin-conjugate (3) whose mitosane moiety is linked to a N-methylpyrrole carboxamide. We found that, overall, both MC and compound 3 show strong similarities regarding their alkylation of DNA, while DMC alkylating behavior is markedly different. To gain further insight into the mode of action of these drugs, we performed high throughput gene expression and gene ontology analysis to identify gene expression and cellular pathways most impacted by each drug treatment in MCF-7 cell lines. We observed that the novel mitomycin derivative (3) specifically causes changes in the expression of genes encoding proteins involved in cell integrity and tissue structure. Further analysis using bioinformatics (IPA) indicated that the new derivative (3) displays a stronger downregulation of major signaling networks that regulate the cell cycle, DNA damage response and cell proliferation when compared to MC and DMC. Collectively, these findings demonstrate that cytotoxic mechanisms of all three drugs are complex and are not solely related to their crosslinking abilities or the structure of the ICLs they produce.
Collapse
Affiliation(s)
- Shu-Yuan Cheng
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, 10016, United States
| | - Lissette Delgado-Cruzata
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Cristina C. Clement
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York, NY, 10065, United States
| | - Owen Zacarias
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Marta Concheiro-Guisan
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Nicholas Towler
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Timothy Snyder
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Maggie Zheng
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Nickolas Almodovar
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Christina Gonzalez
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Marian Romaine
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Anne-Marie Sapse
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
| | - Elise Champeil
- Department of Sciences, John Jay College of Criminal Justice, New York, New York, 10019, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States
| |
Collapse
|
9
|
Assessment of the genotoxic potential of three novel composite nanomaterials using human lymphocytes and the fruit fly Drosophila melanogaster as model systems. CHEMICAL ENGINEERING JOURNAL ADVANCES 2022. [DOI: 10.1016/j.ceja.2021.100230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
10
|
Yamamoto H, Shibuya K, Fukushima T, Hashizume T. Effects of antioxidant capacity on micronucleus induction by cigarette smoke in mammalian cells. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 873:503427. [PMID: 35094812 DOI: 10.1016/j.mrgentox.2021.503427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 06/14/2023]
Abstract
We have compared micronucleus (MN) induction by cigarette smoke in the L5178Y, TK6, and CHL/IU cell lines. The test sample was total particulate matter of 3R4F reference cigarette smoke, suspended in DMSO. After 3-h treatment, with or without a rat liver S9 metabolic activation system, followed by 24-h recovery, dose-dependent MN increases were seen in all cell lines. However, CHL/IU and TK6 cells were more resistant than L5178Y cells (comparison by Benchmark Doses with PROAST software). 3R4F smoke generates reactive oxygen species (ROS). Therefore, we explored the relationship between the sensitivities to 3R4F smoke and the antioxidant capacities of the cell lines. While the total antioxidant capacities were not significantly different among the cell lines, cellular glutathione (GSH) was higher in CHL/IU cells than in L5178Y cells. Pretreatment of CHL/IU cells with a GSH precursor, N-acetylcysteine (NAC), reduced the genotoxicity/cytotoxicity of 3R4F, whereas an inhibitor of GSH biosynthesis, buthionine sulfoximine (BSO), enhanced it. The effects of NAC and BSO were also seen after treatment with allyl isothiocyanate, a ROS-generating chemical, but not with mitomycin C, a ROS-independent genotoxicant. Pretreatment with NAC increased cellular thiol levels. From the present results, the genotoxicity and cytotoxicity of cigarette smoke differs among these cell lines in a manner that may be related to their antioxidant thiol levels.
Collapse
Affiliation(s)
- Haruna Yamamoto
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| | - Kaori Shibuya
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| | - Toshiro Fukushima
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| | - Tsuneo Hashizume
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| |
Collapse
|
11
|
Joseph AM, Daw S, Sadhir I, Badrinarayanan A. Coordination between nucleotide excision repair and specialized polymerase DnaE2 action enables DNA damage survival in non-replicating bacteria. eLife 2021; 10:e67552. [PMID: 33856342 PMCID: PMC8102061 DOI: 10.7554/elife.67552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Translesion synthesis (TLS) is a highly conserved mutagenic DNA lesion tolerance pathway, which employs specialized, low-fidelity DNA polymerases to synthesize across lesions. Current models suggest that activity of these polymerases is predominantly associated with ongoing replication, functioning either at or behind the replication fork. Here we provide evidence for DNA damage-dependent function of a specialized polymerase, DnaE2, in replication-independent conditions. We develop an assay to follow lesion repair in non-replicating Caulobacter and observe that components of the replication machinery localize on DNA in response to damage. These localizations persist in the absence of DnaE2 or if catalytic activity of this polymerase is mutated. Single-stranded DNA gaps for SSB binding and low-fidelity polymerase-mediated synthesis are generated by nucleotide excision repair (NER), as replisome components fail to localize in the absence of NER. This mechanism of gap-filling facilitates cell cycle restoration when cells are released into replication-permissive conditions. Thus, such cross-talk (between activity of NER and specialized polymerases in subsequent gap-filling) helps preserve genome integrity and enhances survival in a replication-independent manner.
Collapse
Affiliation(s)
- Asha Mary Joseph
- National Centre for Biological Sciences - Tata Institute of Fundamental ResearchBangaloreIndia
| | - Saheli Daw
- National Centre for Biological Sciences - Tata Institute of Fundamental ResearchBangaloreIndia
| | - Ismath Sadhir
- National Centre for Biological Sciences - Tata Institute of Fundamental ResearchBangaloreIndia
- Max Planck Institute for Terrestrial Microbiology, LOEWE Centre for Synthetic Microbiology (SYNMIKRO)MarburgGermany
| | - Anjana Badrinarayanan
- National Centre for Biological Sciences - Tata Institute of Fundamental ResearchBangaloreIndia
| |
Collapse
|
12
|
Wolters JEJ, van Mechelen RJS, Al Majidi R, Pinchuk L, Webers CAB, Beckers HJM, Gorgels TGMF. History, presence, and future of mitomycin C in glaucoma filtration surgery. Curr Opin Ophthalmol 2021; 32:148-159. [PMID: 33315724 DOI: 10.1097/icu.0000000000000729] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Mitomycin C (MMC) is an alkylating agent with extraordinary ability to crosslink DNA, preventing DNA synthesis. By this virtue, MMC is an important antitumor drug. In addition, MMC has become the gold standard medication for glaucoma filtration surgery (GFS). This eye surgery creates a passage for drainage of aqueous humor (AqH) out of the eye into the sub-Tenon's space with the aim of lowering the intraocular pressure. A major cause of failure of this operation is fibrosis and scarring in the sub-Tenon's space, which will restrict AqH outflow. Intraoperative application of MMC during GFS has increased GFS success rate, presumably mainly by reducing fibrosis after GFS. However, still 10% of glaucoma surgeries fail within the first year. RECENT FINDINGS In this review, we evaluate risks and benefits of MMC as an adjuvant for GFS. In addition, we discuss possible improvements of its use by adjusting dose and method of administration. SUMMARY One way of improving GFS outcome is to prolong MMC delivery by using a drug delivery system.
Collapse
Affiliation(s)
- Jarno E J Wolters
- University Eye Clinic Maastricht, Maastricht University Medical Centre + (MUMC+), Maastricht
- Chemelot Institute for Science and Technology (InSciTe), Geleen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Ralph J S van Mechelen
- University Eye Clinic Maastricht, Maastricht University Medical Centre + (MUMC+), Maastricht
- Chemelot Institute for Science and Technology (InSciTe), Geleen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Rana Al Majidi
- University Eye Clinic Maastricht, Maastricht University Medical Centre + (MUMC+), Maastricht
- Chemelot Institute for Science and Technology (InSciTe), Geleen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Leonard Pinchuk
- InnFocus, Inc., a Santen Company
- Ophthalmic Biophysics Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Carroll A B Webers
- University Eye Clinic Maastricht, Maastricht University Medical Centre + (MUMC+), Maastricht
- Chemelot Institute for Science and Technology (InSciTe), Geleen
| | - Henny J M Beckers
- University Eye Clinic Maastricht, Maastricht University Medical Centre + (MUMC+), Maastricht
- Chemelot Institute for Science and Technology (InSciTe), Geleen
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht, Maastricht University Medical Centre + (MUMC+), Maastricht
- Chemelot Institute for Science and Technology (InSciTe), Geleen
| |
Collapse
|
13
|
Roske JJ, Liu S, Loll B, Neu U, Wahl MC. A skipping rope translocation mechanism in a widespread family of DNA repair helicases. Nucleic Acids Res 2021; 49:504-518. [PMID: 33300032 PMCID: PMC7797055 DOI: 10.1093/nar/gkaa1174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Mitomycin repair factor A represents a family of DNA helicases that harbor a domain of unknown function (DUF1998) and support repair of mitomycin C-induced DNA damage by presently unknown molecular mechanisms. We determined crystal structures of Bacillus subtilis Mitomycin repair factor A alone and in complex with an ATP analog and/or DNA and conducted structure-informed functional analyses. Our results reveal a unique set of auxiliary domains appended to a dual-RecA domain core. Upon DNA binding, a Zn2+-binding domain, encompassing the domain of unknown function, acts like a drum that rolls out a canopy of helicase-associated domains, entrapping the substrate and tautening an inter-domain linker across the loading strand. Quantification of DNA binding, stimulated ATPase and helicase activities in the wild type and mutant enzyme variants in conjunction with the mode of coordination of the ATP analog suggest that Mitomycin repair factor A employs similar ATPase-driven conformational changes to translocate on DNA, with the linker ratcheting through the nucleotides like a 'skipping rope'. The electrostatic surface topology outlines a likely path for the displaced DNA strand. Our results reveal unique molecular mechanisms in a widespread family of DNA repair helicases linked to bacterial antibiotics resistance.
Collapse
Affiliation(s)
- Johann J Roske
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustraβe 6, D-14195 Berlin, Germany
| | - Sunbin Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustraβe 6, D-14195 Berlin, Germany
| | - Bernhard Loll
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustraβe 6, D-14195 Berlin, Germany
| | - Ursula Neu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Biochemistry of Viruses, Takustraβe 6, D-14195 Berlin, Germany
| | - Markus C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustraβe 6, D-14195 Berlin, Germany.,Helmholtz-Zentrum Berlin für Materialien und Energie, Macromolecular Crystallography, Albert-Einstein-Straße 15, D-12489 Berlin, Germany
| |
Collapse
|
14
|
Aguilar W, Zacarias O, Romaine M, Proni G, Petrovic AG, Abzalimov R, Paz MM, Champeil E. Synthesis of Oligonucleotides containing the cis-Interstrand Crosslink Produced by Mitomycins in their Reaction with DNA. Chemistry 2020; 26:12570-12578. [PMID: 32574396 PMCID: PMC7681910 DOI: 10.1002/chem.202002452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Indexed: 01/25/2023]
Abstract
Mitomycin C (MC) an antitumor drug and decarbamoylmitomycin C (DMC), a derivative of MC lacking the carbamoyl moiety, are DNA alkylating agents which can form DNA interstrand crosslinks (ICLs) between deoxyguanosine residues located on opposing DNA strands. MC forms primarily deoxyguanosine adducts with a 1"-R stereochemistry at the guanine-mitosene bond (1"-α, trans) whereas DMC forms mainly adducts with a 1"-S stereochemistry (1"-β, cis). The crosslinking reaction is diastereospecific: trans-crosslinks are formed exclusively at CpG sequences, while cis-crosslinks are formed only at GpC sequences. Until now, oligonucleotides containing 1"-β-deoxyguanosine adducts or ICL at a specific site could not be synthesized, thus limiting the investigation of the role played by the stereochemical configuration at C1'' in the toxicity of these compounds. Here, a novel biomimetic synthesis to access these substrates is presented. Structural proof of the adducted oligonucleotides and ICL were provided by enzymatic digestion to nucleosides, high resolution mass spectral analysis, CD spectroscopy and UV melting temperature studies. Finally, a virtual model of the 25-mer 1"-β ICL synthesized was created to explore the conformational space and structural features of the crosslinked duplex.
Collapse
Affiliation(s)
- William Aguilar
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Owen Zacarias
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Marian Romaine
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Gloria Proni
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Ana G Petrovic
- New York Institute of Technology, 1855 Broadway, EGGC 405A, New York, NY, 10023, USA
| | - Rinat Abzalimov
- City University of New York, Advanced Research Center, 85 St Nicholas Terrace, New York, NY, 10031, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA
| | - Manuel M Paz
- Departamento de Química Orgánica, Facultad de Química, Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña, 15782, Spain
| | - Elise Champeil
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA
| |
Collapse
|
15
|
Stern HR, Sefcikova J, Chaparro VE, Beuning PJ. Mammalian DNA Polymerase Kappa Activity and Specificity. Molecules 2019; 24:E2805. [PMID: 31374881 PMCID: PMC6695781 DOI: 10.3390/molecules24152805] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 12/31/2022] Open
Abstract
DNA polymerase (pol) kappa is a Y-family translesion DNA polymerase conserved throughout all domains of life. Pol kappa is special6 ized for the ability to copy DNA containing minor groove DNA adducts, especially N2-dG adducts, as well as to extend primer termini containing DNA damage or mismatched base pairs. Pol kappa generally cannot copy DNA containing major groove modifications or UV-induced photoproducts. Pol kappa can also copy structured or non-B-form DNA, such as microsatellite DNA, common fragile sites, and DNA containing G quadruplexes. Thus, pol kappa has roles both in maintaining and compromising genomic integrity. The expression of pol kappa is altered in several different cancer types, which can lead to genome instability. In addition, many cancer-associated single-nucleotide polymorphisms have been reported in the POLK gene, some of which are associated with poor survival and altered chemotherapy response. Because of this, identifying inhibitors of pol kappa is an active area of research. This review will address these activities of pol kappa, with a focus on lesion bypass and cellular mutagenesis.
Collapse
Affiliation(s)
- Hannah R Stern
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Jana Sefcikova
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Victoria E Chaparro
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Penny J Beuning
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Burby PE, Simmons LA. A bacterial DNA repair pathway specific to a natural antibiotic. Mol Microbiol 2018; 111:338-353. [PMID: 30379365 DOI: 10.1111/mmi.14158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2018] [Indexed: 12/17/2022]
Abstract
All organisms possess DNA repair pathways that are used to maintain the integrity of their genetic material. Although many DNA repair pathways are well understood, new pathways continue to be discovered. Here, we report an antibiotic specific DNA repair pathway in Bacillus subtilis that is composed of a previously uncharacterized helicase (mrfA) and exonuclease (mrfB). Deletion of mrfA and mrfB results in sensitivity to the DNA damaging agent mitomycin C, but not to any other type of DNA damage tested. We show that MrfAB function independent of canonical nucleotide excision repair, forming a novel excision repair pathway. We demonstrate that MrfB is a metal-dependent exonuclease and that the N-terminus of MrfB is required for interaction with MrfA. We determined that MrfAB failed to unhook interstrand cross-links in vivo, suggesting that MrfAB are specific to the monoadduct or the intrastrand cross-link. A phylogenetic analysis uncovered MrfAB homologs in diverse bacterial phyla, and cross-complementation indicates that MrfAB function is conserved in closely related species. B. subtilis is a soil dwelling organism and mitomycin C is a natural antibiotic produced by the soil bacterium Streptomyces lavendulae. The specificity of MrfAB suggests that these proteins are an adaptation to environments with mitomycin producing bacteria.
Collapse
Affiliation(s)
- Peter E Burby
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lyle A Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
17
|
Aguilar W, Paz MM, Vargas A, Zheng M, Cheng SY, Champeil E. Interdependent Sequence Selectivity and Diastereoselectivity in the Alkylation of DNA by Decarbamoylmitomycin C. Chemistry 2018; 24:13278-13289. [PMID: 29958326 PMCID: PMC7152928 DOI: 10.1002/chem.201802038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/27/2018] [Indexed: 02/01/2023]
Abstract
Mitomycin C (MC), an antitumor drug, and decarbamoylmitomycin C (DMC), a derivative of MC, alkylate DNA and form deoxyguanosine monoadducts and interstrand crosslinks (ICLs). Interestingly, in mammalian culture cells, MC forms primarily deoxyguanosine adducts with a 1"-R stereochemistry at the guanine-mitosene bond (1"-α) whereas DMC forms mainly adducts with a 1"-S stereochemistry (1"-β). The molecular basis for the stereochemical configuration exhibited by DMC has been investigated using biomimetic synthesis. Here, we present the results of our studies on the monoalkylation of DNA by DMC. We show that the formation of 1"-β-deoxyguanosine adducts requires bifunctional reductive activation of DMC, and that monofunctional activation only produces 1"-α-adducts. The stereochemistry of the deoxyguanosine adducts formed is also dependent on the regioselectivity of DNA alkylation and on the overall DNA CG content. Additionally, we found that temperature plays a determinant role in the regioselectivity of duplex DNA alkylation by mitomycins: At 0 °C, both deoxyadenosine (dA) and deoxyguanosine (dG) alkylation occur whereas at 37 °C, mitomycins alkylate dG preferentially. The new reaction protocols developed in our laboratory to investigate DMC-DNA alkylation raise the possibility that oligonucleotides containing DMC 1"-β-deoxyguanosine adducts at a specific site may be synthesized by a biomimetic approach.
Collapse
Affiliation(s)
- William Aguilar
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Manuel M Paz
- Departamento de Química Orgánica, Facultade de Química, Universidade de Santiago de Compostela, 15782, Santiago, de Compostela, Spain
| | - Anayatzinc Vargas
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Maggie Zheng
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Shu-Yuan Cheng
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Elise Champeil
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City, University of New York, New York, NY, 10016, USA
| |
Collapse
|
18
|
Aguilar W, Paz MM, Vargas A, Clement CC, Cheng SY, Champeil E. Sequence-Dependent Diastereospecific and Diastereodivergent Crosslinking of DNA by Decarbamoylmitomycin C. Chemistry 2018; 24:6030-6035. [PMID: 29504661 PMCID: PMC7046179 DOI: 10.1002/chem.201705771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Indexed: 11/08/2022]
Abstract
Mitomycin C (MC), a potent antitumor drug, and decarbamoylmitomycin C (DMC), a derivative lacking the carbamoyl group, form highly cytotoxic DNA interstrand crosslinks. The major interstrand crosslink formed by DMC is the C1'' epimer of the major crosslink formed by MC. The molecular basis for the stereochemical configuration exhibited by DMC was investigated using biomimetic synthesis. The formation of DNA-DNA crosslinks by DMC is diastereospecific and diastereodivergent: Only the 1''S-diastereomer of the initially formed monoadduct can form crosslinks at GpC sequences, and only the 1''R-diastereomer of the monoadduct can form crosslinks at CpG sequences. We also show that CpG and GpC sequences react with divergent diastereoselectivity in the first alkylation step: 1"S stereochemistry is favored at GpC sequences and 1''R stereochemistry is favored at CpG sequences. Therefore, the first alkylation step results, at each sequence, in the selective formation of the diastereomer able to generate an interstrand DNA-DNA crosslink after the "second arm" alkylation. Examination of the known DNA adduct pattern obtained after treatment of cancer cell cultures with DMC indicates that the GpC sequence is the major target for the formation of DNA-DNA crosslinks in vivo by this drug.
Collapse
Affiliation(s)
- William Aguilar
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Manuel M Paz
- Departamento de Química Orgánica, Facultade de Química, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Anayatzinc Vargas
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Cristina C Clement
- Pathology Department, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Chemistry, Lehman College of the City University of New York, Bronx, New York, 10468, USA
| | - Shu-Yuan Cheng
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
| | - Elise Champeil
- Science Department, John Jay College of Criminal Justice, 524 West 59th street, New York, NY, 10019, USA
- The Graduate Center of the, City University of New York, New York, NY, 10016, USA
| |
Collapse
|
19
|
Pandey S, Kirti A, Kumar A, Rajaram H. The SbcC and SbcD homologs of the cyanobacterium Anabaena sp. strain PCC7120 (Alr3988 and All4463) contribute independently to DNA repair. Funct Integr Genomics 2018. [DOI: 10.1007/s10142-018-0599-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
20
|
Kumari S, Nayak G, Lukose ST, Kalthur SG, Bhat N, Hegde AR, Mutalik S, Kalthur G, Adiga SK. Indian propolis ameliorates the mitomycin C-induced testicular toxicity by reducing DNA damage and elevating the antioxidant activity. Biomed Pharmacother 2017; 95:252-263. [PMID: 28846983 DOI: 10.1016/j.biopha.2017.08.065] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 01/06/2023] Open
Abstract
Development of excellent curative therapy for most of the malignancies has resulted in a growing population of cancer survivors who are at increased risk for a variety of health problems including infertility. Therefore, fertility preservation has become an important issue during cancer treatment in recent years. Combination therapy with natural agents such as vitamins, antioxidants, dietary supplements, and plant products are considered as an attractive option to mitigate normal tissue toxicity imparted by chemotherapy. The aim of the present study was to explore the beneficial effect of hydroethanolic extract of Indian propolis (HEIP) on mitigating mitomycin C (MMC)-induced testicular damage and its mechanism of action. Healthy adult male mice were injected intraperitoneally with saline, MMC, HEIP and HEIP followed by MMC after 1h. The animals were dissected at 35days after various treatments to analyze testicular function. MMC administration resulted in significant reduction in testicular function in a dose-dependent manner at 35days after treatment which significantly improved by HEIP pre-treatment. At 24h after treatment, MMC induced significant increase in oxidative stress, γ-H2AX foci and expression of RAD51 and KU80 in testicular cells. Prior treatment with HEIP decreased the oxidative stress, reduced DNA damage and restored the testicular testosterone and inhibin B level. In conclusion, co-administration of Indian propolis extract may play a promising beneficial role in fertility preservation of males undergoing chemotherapy.
Collapse
Affiliation(s)
- Sandhya Kumari
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal University, Manipal, 576 104, Karnataka, India
| | - Guruprasad Nayak
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal University, Manipal, 576 104, Karnataka, India
| | - Sonu T Lukose
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal University, Manipal, 576 104, Karnataka, India
| | - Sneha Guruprasad Kalthur
- Department of Anatomy, Kasturba Medical College, Manipal University, Manipal, 576 104, Karnataka, India
| | - Nandini Bhat
- Department of Anatomy, Kasturba Medical College, Manipal University, Manipal, 576 104, Karnataka, India
| | - Aswathi R Hegde
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, 576 104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, 576 104, Karnataka, India
| | - Guruprasad Kalthur
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal University, Manipal, 576 104, Karnataka, India.
| | - Satish Kumar Adiga
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal University, Manipal, 576 104, Karnataka, India
| |
Collapse
|
21
|
Bruzaca EES, Lopes IC, Silva EHC, Carvalho PAV, Tanaka AA. Electrochemical oxidation of the antitumor antibiotic mitomycin C and in situ evaluation of its interaction with DNA using a DNA-electrochemical biosensor. Microchem J 2017. [DOI: 10.1016/j.microc.2017.03.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
22
|
Gorniewska AM, Kluzek K, Gackowska L, Kubiszewska I, Zdzienicka MZ, Bialkowska A. Distinct cellular phenotype linked to defective DNA interstrand crosslink repair and homologous recombination. Mol Med Rep 2017. [PMID: 28627616 PMCID: PMC5561886 DOI: 10.3892/mmr.2017.6781] [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] [Indexed: 01/22/2023] Open
Abstract
Repair of DNA interstrand crosslinks (ICLs) predominantly involves the Fanconi anemia (FA) pathway and homologous recombination (HR). The HR repair system eliminates DNA double strand breaks (DSBs) that emerge during ICLs removal. The current study presents a novel cell line, CL-V8B, representing a new complementation group of Chinese hamster cell mutants hypersensitive to DNA crosslinking factors. CL-V8B exhibits increased sensitivity to various DNA-damaging agents, including compounds leading to DSBs formation (bleomycin and 6-thioguanine), and is extremely sensitive to poly (ADP-ribose) polymerase inhibitor (>400-fold), which is typical for HR-defective cells. In addition, this cell line exhibits a reduced number of spontaneous and induced sister chromatid exchanges, which suggests likely impairment of HR in CL-V8B cells. However, in contrast to other known HR mutants, CL-V8B cells do not show defects in Rad51 foci induction, but only slight alterations in the focus formation kinetics. CL-V8B is additionally characterized by a considerable chromosomal instability, as indicated by a high number of spontaneous and MMC-induced chromosomal aberrations, and a twice as large proportion of cells with abnormal centrosomes than that in the wild type cell line. The molecular defect present in CL-V8B does not affect the efficiency and stabilization of replication forks. However, stalling of the forks in response to replication stress is observed relatively rarely, which suggests an impairment of a signaling mechanism. Exposure of CL-V8B to crosslinking agents results in S-phase arrest (as in the wild type cells), but also in larger proportion of G2/M-phase cells and apoptotic cells. CL-V8B exhibits similarities to HR- and/or FA-defective Chinese hamster mutants sensitive to DNA crosslinking agents. However, the unique phenotype of this new mutant implies that it carries a defect of a yet unidentified gene involved in the repair of ICLs.
Collapse
Affiliation(s)
- Aleksandra M Gorniewska
- Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz 85‑094, Poland
| | - Katarzyna Kluzek
- Department of Human Molecular Genetics, Adam Mickiewicz University, Poznan 61‑614, Poland
| | - Lidia Gackowska
- Department of Immunology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz 85‑094, Poland
| | - Izabela Kubiszewska
- Department of Immunology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz 85‑094, Poland
| | - Malgorzata Z Zdzienicka
- Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz 85‑094, Poland
| | - Aneta Bialkowska
- Innovative Medical Forum, Franciszek Lukaszczyk Oncology Center, Bydgoszcz 85‑796, Poland
| |
Collapse
|
23
|
Stornetta A, Zimmermann M, Cimino GD, Henderson PT, Sturla SJ. DNA Adducts from Anticancer Drugs as Candidate Predictive Markers for Precision Medicine. Chem Res Toxicol 2017; 30:388-409. [PMID: 27936622 PMCID: PMC5379252 DOI: 10.1021/acs.chemrestox.6b00380] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Indexed: 01/23/2023]
Abstract
Biomarker-driven drug selection plays a central role in cancer drug discovery and development, and in diagnostic strategies to improve the use of traditional chemotherapeutic drugs. DNA-modifying anticancer drugs are still used as first line medication, but drawbacks such as resistance and side effects remain an issue. Monitoring the formation and level of DNA modifications induced by anticancer drugs is a potential strategy for stratifying patients and predicting drug efficacy. In this perspective, preclinical and clinical data concerning the relationship between drug-induced DNA adducts and biological response for platinum drugs and combination therapies, nitrogen mustards and half-mustards, hypoxia-activated drugs, reductase-activated drugs, and minor groove binding agents are presented and discussed. Aspects including measurement strategies, identification of adducts, and biological factors that influence the predictive relationship between DNA modification and biological response are addressed. A positive correlation between DNA adduct levels and response was observed for the majority of the studies, demonstrating the high potential of using DNA adducts from anticancer drugs as mechanism-based biomarkers of susceptibility, especially as bioanalysis approaches with higher sensitivity and throughput emerge.
Collapse
Affiliation(s)
- Alessia Stornetta
- Department
of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| | - Maike Zimmermann
- Department
of Internal Medicine, Division of Hematology and Oncology and the
UC Davis Comprehensive Cancer Center, University
of California Davis, 4501 X Street, Sacramento, California 95655, United States
- Accelerated
Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, California 95618, United States
| | - George D. Cimino
- Accelerated
Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, California 95618, United States
| | - Paul T. Henderson
- Department
of Internal Medicine, Division of Hematology and Oncology and the
UC Davis Comprehensive Cancer Center, University
of California Davis, 4501 X Street, Sacramento, California 95655, United States
- Accelerated
Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, California 95618, United States
| | - Shana J. Sturla
- Department
of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| |
Collapse
|
24
|
Matsumoto T, Nishikawa T, Furukawa A, Itano S, Tamura Y, Hasei T, Watanabe T. Antimutagenic Effects of Polymethoxy Flavonoids of Citrus unshiu. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701200108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Citrus fruits have been used as edible fruit and traditional medicine for various diseases such as cancer. In the courses of our study to find antimutagens, we have found that the ethanolic extract of the peel of Citrus unshiu Marc showed antimutagenic effects against several mutagens in the Ames test using Salmonella typhimurium TA98 strain. Three polymethoxy flavonoids, nobiletin, 3,5,6,7,8,3′,4′-heptamethoxyflavone, and tangeretin, were identified in the extract as major constituents. These three polymethoxy flavonoids showed antimutagenic effects in the Ames test in vitro and in the micronucleus test in vivo.
Collapse
Affiliation(s)
- Takahiro Matsumoto
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Taisuke Nishikawa
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Ayano Furukawa
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Saki Itano
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Yuka Tamura
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Tomohiro Hasei
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Tetsushi Watanabe
- Department of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| |
Collapse
|
25
|
Cheng SY, Seo J, Huang BT, Napolitano T, Champeil E. Mitomycin C and decarbamoyl mitomycin C induce p53-independent p21WAF1/CIP1 activation. Int J Oncol 2016; 49:1815-1824. [PMID: 27666201 PMCID: PMC5063421 DOI: 10.3892/ijo.2016.3703] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022] Open
Abstract
Mitomycin C (MC), a commonly used anticancer drug, induces DNA damage via DNA alkylation. Decarbamoyl mitomycin C (DMC), another mitomycin lacking the carbamate at C10, generates similar lesions as MC. Interstrand cross-links (ICLs) are believed to be the lesions primarily responsible for the cytotoxicity of MC and DMC. The major ICL generated by MC (α-ICL) has a trans stereochemistry at the guanine-drug linkage whereas the major ICL from DMC (β-ICL) has the opposite, cis, stereochemistry. In addition, DMC can provoke strong p53-independent cell death. Our hypothesis is that the stereochemistry of the major unique β-ICL generated by DMC is responsible for this p53-independent cell death signaling. p53 gene is inactively mutated in more than half of human cancers. p21WAF1/CIP1 known as a major effector of p53 is involved in p53-dependent and -independent control of cell proliferation and death. This study revealed the role of p21WAF1/CIP1 on MC and DMC triggered cell damage. MCF-7 (p53-proficient) and K562 (p53-deficient) cells were used. Cell cycle distributions were shifted to the G1/S phase in MCF-7 treated with MC and DMC, but were shifted to the S phase in K562. p21WAF1/CIP1 activation was observed in both cells treated with MC and DMC, and DMC triggered more significant activation. Knocking down p53 in MCF-7 did not attenuate MC and DMC induced p21WAF1/CIP1 activation. The α-ICL itself was enough to cause p21WAF1/CIP1 activation.
Collapse
Affiliation(s)
- Shu-Yuan Cheng
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, NY 10019, USA
| | - Jiwon Seo
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, NY 10019, USA
| | - Bik Tzu Huang
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, NY 10019, USA
| | - Tanya Napolitano
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, NY 10019, USA
| | - Elise Champeil
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, NY 10019, USA
| |
Collapse
|
26
|
Hovhannisyan G, Aroutiounian R, Babayan N, Harutyunyan T, Liehr T. Comparative analysis of individual chromosome involvement in micronuclei induced by mitomycin C and bleomycin in human leukocytes. Mol Cytogenet 2016; 9:49. [PMID: 27330564 PMCID: PMC4915088 DOI: 10.1186/s13039-016-0258-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/15/2016] [Indexed: 12/18/2022] Open
Abstract
Background Micronucleus (MN) assay is a well standardized approach for evaluation of clastogenic/aneugenic effects of mutagens. Fluorescence in situ hybridization (FISH) is successfully used to characterize the chromosomal content of MN. However, the relationships between nuclear positioning, length, and gene density of individual chromosomes and their involvement in MN induced by different mutagens have not been clearly defined. Results Chromosomal content of MN was characterized in human leukocytes treated with mitomycin C (MMC) and bleomycin (BLM) by FISH using centromeric (cep) and whole-chromosome painting (wcp) probes. Involvement of chromosomes 8, 15 and 20 in MMC-induced and chromosomes 1, 9 and 16 in BLM-induced MN was studied, and correlated with chromosome size, gene density and interphase position. The results obtained were analyzed together with previous own data on the frequencies of inclusion of chromosomes 3, 4, 6, 7, 9, 16, 17, 18, and X in MMC-induced MN. It could be shown that MMC- and BLM-induced MN could contain material derived from all chromosomes investigated. Involvement of whole chromosomes 8, 15 and 20 in MMC-induced MN negatively correlated with gene density; however, analysis together with earlier studied chromosomes did not confirm this correlation. Inclusion of chromosomes 8, 15 and 20 in MMC-induced MN does not depend on their size and interphase position; the same result was found for the twelve overall analyzed chromosomes. In BLM-treated cells significant correlation between frequencies of involvement of chromosomes 1, 9 and 16 in MN and their size was found. Conclusions Our results clearly revealed that BLM differs from MMC with respect to the distribution of induced chromosome damage and MN formation. Thus, DNA-damaging agents with diverse mechanism of action induce qualitatively different MN with regard to their chromosomal composition. Also this study demonstrates the utility of combined sequential application of cep and wcp probes for efficient detection of MN chromosomal content in terms of centric and acentric fragments.
Collapse
Affiliation(s)
- Galina Hovhannisyan
- Department of Genetics and Cytology, Faculty of Biology, Yerevan State University, 1 Alex Manoogian, 0025 Yerevan, Armenia
| | - Rouben Aroutiounian
- Department of Genetics and Cytology, Faculty of Biology, Yerevan State University, 1 Alex Manoogian, 0025 Yerevan, Armenia
| | - Nelly Babayan
- Department of Genetics and Cytology, Faculty of Biology, Yerevan State University, 1 Alex Manoogian, 0025 Yerevan, Armenia ; Institute of Molecular Biology, National Academy of Sciences, 7 Hasratyan, 0014 Yerevan, Armenia
| | - Tigran Harutyunyan
- Department of Genetics and Cytology, Faculty of Biology, Yerevan State University, 1 Alex Manoogian, 0025 Yerevan, Armenia
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Kollegiengasse 10, D-07743 Jena, Germany
| |
Collapse
|
27
|
Bose A, Surugihalli C, Pande P, Champeil E, Basu AK. Comparative Error-Free and Error-Prone Translesion Synthesis of N(2)-2'-Deoxyguanosine Adducts Formed by Mitomycin C and Its Metabolite, 2,7-Diaminomitosene, in Human Cells. Chem Res Toxicol 2016; 29:933-9. [PMID: 27082015 PMCID: PMC4871107 DOI: 10.1021/acs.chemrestox.6b00087] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Indexed: 11/28/2022]
Abstract
Mitomycin C (MC) is a cytotoxic and mutagenic antitumor agent that alkylates DNA upon reductive activation. 2,7-Diaminomitosene (2,7-DAM) is a major metabolite of MC in tumor cells, which also alkylates DNA. MC forms seven DNA adducts, including monoadducts and inter- and intrastrand cross-links, whereas 2,7-DAM forms two monoadducts. Herein, the biological effects of the dG-N(2) adducts formed by MC and 2,7-DAM have been compared by constructing single-stranded plasmids containing these adducts and replicating them in human embryonic kidney 293T cells. Translesion synthesis (TLS) efficiencies of dG-N(2)-MC and dG-N(2)-2,7-DAM were 38 ± 3 and 27 ± 3%, respectively, compared to that of a control plasmid. This indicates that both adducts block DNA synthesis and that dG-N(2)-2,7-DAM is a stronger replication block than dG-N(2)-MC. TLS of each adducted construct was reduced upon siRNA knockdown of pol η, pol κ, or pol ζ. For both adducts, the most significant reduction occurred with knockdown of pol κ, which suggests that pol κ plays a major role in TLS of these dG-N(2) adducts. Analysis of the progeny showed that both adducts were mutagenic, and the mutation frequencies (MF) of dG-N(2)-MC and dG-N(2)-2,7-DAM were 18 ± 3 and 10 ± 1%, respectively. For both adducts, the major type of mutation was G → T transversions. Knockdown of pol η and pol ζ reduced the MF of dG-N(2)-MC and dG-N(2)-2,7-DAM, whereas knockdown of pol κ increased the MF of these adducts. This suggests that pol κ predominantly carries out error-free TLS, whereas pol η and pol ζ are involved in error-prone TLS. The largest reduction in MF by 78 and 80%, respectively, for dG-N(2)-MC and dG-N(2)-2,7-DAM constructs occurred when pol η, pol ζ, and Rev1 were simultaneously knocked down. This result strongly suggests that, unlike pol κ, these three TLS polymerases cooperatively perform the error-prone TLS of these adducts.
Collapse
Affiliation(s)
- Arindam Bose
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Chaitra Surugihalli
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Paritosh Pande
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Elise Champeil
- Department
of Science, John Jay College of Criminal
Justice, New York, New York 10019, United
States
| | - Ashis K. Basu
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
28
|
Xiao G, Kue P, Bhosle R, Bargonetti J. Decarbamoyl mitomycin C (DMC) activates p53-independent ataxia telangiectasia and rad3 related protein (ATR) chromatin eviction. Cell Cycle 2015; 14:744-54. [PMID: 25565400 PMCID: PMC4418290 DOI: 10.1080/15384101.2014.997517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Interstrand crosslinks induce DNA replication fork stalling that in turn activates the ATR-dependent checkpoint and DNA repair on nuclear chromatin. Mitomycin C (MC) and Decarbamoyl Mitomycin C (DMC) induce different types of DNA crosslinks with DMC being a more cytotoxic agent. We previously reported that the novel DMC induced β–interstrand DNA crosslinks induce a p53-independent form of cell death. The p53-independent DMC cytotoxicity associates with the activation, and subsequent depletion, of Chk1. In this study we further dissect the novel DMC signal transduction pathway and asked how it influences chromatin-associated proteins. We found that treatment with DMC, but not MC, stimulated the disassociation of ATR from chromatin and re-localization of ATR to the cytoplasm. The chromatin eviction of ATR was coupled with the formation of nuclear Rad51 foci and the phosphorylation of Chk1. Furthermore, DMC but not MC, activated expression of gadd45α mRNA. Importantly, knocking down p53 via shRNA did not inhibit the DMC-induced disassociation of ATR from chromatin or reduce the activation of transcription of gadd45α. Our results suggest that DMC induces a p53-independent disassociation of ATR from chromatin that facilitates Chk1 checkpoint activation and Rad51 chromatin recruitment. Our findings provide evidence that ATR chromatin eviction in breast cancer cells is an area of study that should be focused on for inducing p53-independent cell death.
Collapse
Key Words
- ATR
- ATR, Ataxia Telangiectasia and Rad3 Related
- Cdc25, cell division cycle 25.
- ChIP, chromatin immunoprecipitation
- Chk1, checkpoint serine/threonine protein kinase 1
- DDR, DNA damage response
- DMC, Decarbamoyl Mitomycin C
- DOX, doxycycline
- HR, homologous recombination
- ICLs, interstrand cross-links
- MC, Mitomycin C
- NER, nuclear excision repair
- PCNA, Proliferating Cell Nuclear Antigen
- RPA, replication protein A
- TNBCs, triple negative breast cancers
- breast cancer
- cell death
- chromatin
- interstrand crosslink
- mitomycin C
- p53
Collapse
Affiliation(s)
- Gu Xiao
- a Department of Biological Sciences; Hunter College and The Graduate Center Biology Program ; City University of New York ; New York , NY USA
| | | | | | | |
Collapse
|
29
|
Giroux X, MacNeill SA. A novel archaeal DNA repair factor that acts with the UvrABC system to repair mitomycin C-induced DNA damage in a PCNA-dependent manner. Mol Microbiol 2015; 99:1-14. [PMID: 26337406 DOI: 10.1111/mmi.13210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2015] [Indexed: 01/11/2023]
Abstract
The sliding clamp proliferating cell nuclear antigen (PCNA) plays a vital role in a number of DNA repair pathways in eukaryotes and archaea by acting as a stable platform onto which other essential protein factors assemble. Many of these proteins interact with PCNA via a short peptide sequence known as a PIP (PCNA interacting protein) motif. Here we describe the identification and functional analysis of a novel PCNA interacting protein NreA that is conserved in the archaea and that has a PIP motif at its C-terminus. Using the genetically tractable euryarchaeon Haloferax volcanii as a model system, we show that the NreA protein is not required for cell viability but that loss of NreA (or replacement of the wild-type protein with a truncated version lacking the C-terminal PIP motif) results in an increased sensitivity to the DNA damaging agent mitomycin C (MMC) that correlates with delayed repair of MMC-induced chromosomal DNA damage monitored by pulsed-field gel electrophoresis. Genetic epistasis analysis in Hfx. volcanii suggests that NreA works together with the UvrABC proteins in repairing DNA damage resulting from exposure to MMC. The wide distribution of NreA family members implies an important role for the protein in DNA damage repair in all archaeal lineages.
Collapse
Affiliation(s)
- Xavier Giroux
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, North Haugh, St Andrews, Fife, KY16 9ST, UK
| | - Stuart A MacNeill
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, North Haugh, St Andrews, Fife, KY16 9ST, UK
| |
Collapse
|
30
|
Vlastos D, Drosopoulou E, Efthimiou I, Gavriilidis M, Panagaki D, Mpatziou K, Kalamara P, Mademtzoglou D, Mavragani-Tsipidou P. Genotoxic and Antigenotoxic Assessment of Chios Mastic Oil by the In Vitro Micronucleus Test on Human Lymphocytes and the In Vivo Wing Somatic Test on Drosophila. PLoS One 2015; 10:e0130498. [PMID: 26110900 PMCID: PMC4482422 DOI: 10.1371/journal.pone.0130498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/19/2015] [Indexed: 12/16/2022] Open
Abstract
Chios mastic oil (CMO), the essential oil derived from Pistacia lentiscus (L.) var. chia (Duham), has generated considerable interest because of its antimicrobial, anticancer, antioxidant and other beneficial properties. In the present study, the potential genotoxic activity of CMO as well as its antigenotoxic properties against the mutagenic agent mitomycin-C (MMC) were evaluated by employing the in vitro Cytokinesis Block MicroNucleus (CBMN) assay and the in vivo Somatic Mutation And Recombination Test (SMART). In the in vitro experiments, lymphocytes were treated with 0.01, 0.05 and 0.10% (v/v) of CMO with or without 0.05 μg/ml MMC, while in the in vivo assay Drosophila larvae were fed with 0.05, 0.10, 0.50 and 1.00% (v/v) of CMO with or without 2.50 μg/ml MMC. CMO did not significantly increase the frequency of micronuclei (MN) or total wing spots, indicating lack of mutagenic or recombinogenic activity. However, the in vitro analysis suggested cytotoxic activity of CMO. The simultaneous administration of MMC with CMO did not alter considerably the frequencies of MMC-induced MN and wing spots showing that CMO doesn't exert antigenotoxic or antirecombinogenic action. Therefore, CMO could be considered as a safe product in terms of genotoxic potential. Even though it could not afford any protection against DNA damage, at least under our experimental conditions, its cytotoxic potential could be of interest.
Collapse
Affiliation(s)
- Dimitris Vlastos
- Department of Environmental and Natural Resources Management, University of Patras, Agrinio, Greece
| | - Elena Drosopoulou
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Efthimiou
- Department of Environmental and Natural Resources Management, University of Patras, Agrinio, Greece
| | - Maximos Gavriilidis
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitra Panagaki
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Krystalenia Mpatziou
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paraskevi Kalamara
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despoina Mademtzoglou
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Penelope Mavragani-Tsipidou
- Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
31
|
Ahmadzai AA, Trevisan J, Pang W, Riding MJ, Strong RJ, Llabjani V, Pant K, Carmichael PL, Scott AD, Martin FL. Classification of agents using Syrian hamster embryo (SHE) cell transformation assay (CTA) with ATR-FTIR spectroscopy and multivariate analysis. Mutagenesis 2015; 30:603-12. [PMID: 25925069 DOI: 10.1093/mutage/gev030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The Syrian hamster embryo (SHE) cell transformation assay (pH 6.7) has a reported sensitivity of 87% and specificity of 83%, and an overall concordance of 85% with in vivo rodent bioassay data. To date, the SHE assay is the only in vitro assay that exhibits multistage carcinogenicity. The assay uses morphological transformation, the first stage towards neoplasm, as an endpoint to predict the carcinogenic potential of a test agent. However, scoring of morphologically transformed SHE cells is subjective. We treated SHE cells grown on low-E reflective slides with 2,6-diaminotoluene, N-nitroso-N-ethylnitroguanidine, N-nitroso-N-methylurea, N-nitroso-N-ethylurea, EDTA, dimethyl sulphoxide (DMSO; vehicle control), methyl methanesulfonate, benzo[e]pyrene, mitomycin C, ethyl methanesulfonate, ampicillin or five different concentrations of benzo[a]pyrene. Macroscopically visible SHE colonies were located on the slides and interrogated using attenuated total reflection Fourier-transform infrared (ATR-FTIR) spectroscopy acquiring five spectra per colony. The acquired IR data were analysed using Fisher's linear discriminant analysis (LDA) followed by principal component analysis (PCA)-LDA cluster vectors to extract major and minor discriminating wavenumbers for each treatment class. Each test agent vs. DMSO and treatment-induced transformed cells vs. corresponding non-transformed were classified by a unique combination of major and minor discriminating wavenumbers. Alterations associated with Amide I, Amide II, lipids and nucleic acids appear to be important in segregation of classes. Our findings suggest that a biophysical approach of ATR-FTIR spectroscopy with multivariate analysis could facilitate a more objective interrogation of SHE cells towards scoring for transformation and ultimately employing the assay for risk assessment of test agents.
Collapse
Affiliation(s)
- Abdullah A Ahmadzai
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Júlio Trevisan
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Weiyi Pang
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Matthew J Riding
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Rebecca J Strong
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Valon Llabjani
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Kamala Pant
- BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and
| | - Paul L Carmichael
- Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Andrew D Scott
- Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| | - Francis L Martin
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Lancaster LA1 4YQ, UK, BioReliance by SAFC, Medical Center Drive, Rockville, MD 20850, USA and Safety and Environmental Assurance Centre, Unilever Colworth Science Park, Bedfordshire, MK44 1LQ, UK
| |
Collapse
|
32
|
Haeger SM, Thompson JJ, Kalra S, Cleaver TG, Merrick D, Wang XJ, Malkoski SP. Smad4 loss promotes lung cancer formation but increases sensitivity to DNA topoisomerase inhibitors. Oncogene 2015; 35:577-586. [PMID: 25893305 PMCID: PMC4615192 DOI: 10.1038/onc.2015.112] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/21/2015] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a common malignancy with a poor prognosis. Despite progress targeting oncogenic drivers, there are no therapies targeting tumor suppressor loss. Smad4 is an established tumor suppressor in pancreatic and colon cancer, however, the consequences of Smad4 loss in lung cancer are largely unknown. We evaluated Smad4 expression in human NSCLC samples and examined Smad4 alterations in large NSCLC datasets and found that reduced Smad4 expression is common in human NSCLC and occurs through a variety of mechanisms including mutation, homozygous deletion, and heterozygous loss. We modeled Smad4 loss in lung cancer by deleting Smad4 in airway epithelial cells and found that Smad4 deletion both initiates and promotes lung tumor development. Interestingly, both Smad4−/− mouse tumors and human NSCLC samples with reduced Smad4 expression demonstrated increased DNA damage while Smad4 knockdown in lung cancer cells reduced DNA repair and increased apoptosis after DNA damage. In addition, Smad4 deficient NSCLC cells demonstrated increased sensitivity to both chemotherapeutics that inhibit DNA topoisomerase and drugs that block double strand DNA break repair by non-homologous end joining. In sum, these studies establish Smad4 as a lung tumor suppressor and suggest that the defective DNA repair phenotype of Smad4 deficient tumors can be exploited by specific therapeutic strategies.
Collapse
Affiliation(s)
- Sarah M Haeger
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Joshua J Thompson
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Sean Kalra
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Timothy G Cleaver
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Daniel Merrick
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Stephen P Malkoski
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO.,Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
33
|
Pomyen Y, Segura M, Ebbels TMD, Keun HC. Over-representation of correlation analysis (ORCA): a method for identifying associations between variable sets. Bioinformatics 2014; 31:102-8. [DOI: 10.1093/bioinformatics/btu589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
34
|
Kristensen TP, Maria Cherian R, Gray FC, MacNeill SA. The haloarchaeal MCM proteins: bioinformatic analysis and targeted mutagenesis of the β7-β8 and β9-β10 hairpin loops and conserved zinc binding domain cysteines. Front Microbiol 2014; 5:123. [PMID: 24723920 PMCID: PMC3972481 DOI: 10.3389/fmicb.2014.00123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 03/10/2014] [Indexed: 11/13/2022] Open
Abstract
The hexameric MCM complex is the catalytic core of the replicative helicase in eukaryotic and archaeal cells. Here we describe the first in vivo analysis of archaeal MCM protein structure and function relationships using the genetically tractable haloarchaeon Haloferax volcanii as a model system. Hfx. volcanii encodes a single MCM protein that is part of the previously identified core group of haloarchaeal MCM proteins. Three structural features of the N-terminal domain of the Hfx. volcanii MCM protein were targeted for mutagenesis: the β7-β8 and β9-β10 β-hairpin loops and putative zinc binding domain. Five strains carrying single point mutations in the β7-β8 β-hairpin loop were constructed, none of which displayed impaired cell growth under normal conditions or when treated with the DNA damaging agent mitomycin C. However, short sequence deletions within the β7-β8 β-hairpin were not tolerated and neither was replacement of the highly conserved residue glutamate 187 with alanine. Six strains carrying paired alanine substitutions within the β9-β10 β-hairpin loop were constructed, leading to the conclusion that no individual amino acid within that hairpin loop is absolutely required for MCM function, although one of the mutant strains displays greatly enhanced sensitivity to mitomycin C. Deletions of two or four amino acids from the β9-β10 β-hairpin were tolerated but mutants carrying larger deletions were inviable. Similarly, it was not possible to construct mutants in which any of the conserved zinc binding cysteines was replaced with alanine, underlining the likely importance of zinc binding for MCM function. The results of these studies demonstrate the feasibility of using Hfx. volcanii as a model system for reverse genetic analysis of archaeal MCM protein function and provide important confirmation of the in vivo importance of conserved structural features identified by previous bioinformatic, biochemical and structural studies.
Collapse
Affiliation(s)
- Tatjana P Kristensen
- Department of Biology, University of Copenhagen, Københavns Biocenter Copenhagen N, Denmark
| | - Reeja Maria Cherian
- Department of Biology, University of Copenhagen, Københavns Biocenter Copenhagen N, Denmark
| | - Fiona C Gray
- Department of Biology, University of Copenhagen, Københavns Biocenter Copenhagen N, Denmark
| | - Stuart A MacNeill
- Department of Biology, University of Copenhagen, Københavns Biocenter Copenhagen N, Denmark ; School of Biology, University of St. Andrews North Haugh, St. Andrews, Fife, UK
| |
Collapse
|
35
|
Cameron RS, Liu C, Pihkala JPS. Myosin 16 levels fluctuate during the cell cycle and are downregulated in response to DNA replication stress. Cytoskeleton (Hoboken) 2013; 70:328-48. [PMID: 23596177 DOI: 10.1002/cm.21109] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 03/27/2013] [Indexed: 01/03/2023]
Abstract
Myosins comprise a highly conserved superfamily of eukaryotic actin-dependent motor proteins implicated in a large repertoire of functions in both the cytoplasm and the nucleus. Class XVI myosin, MYO16, reveals expression in most somatic as well as meiotic cells with prominent localization in the nucleus, excepting the nucleolus; however, the role(s) of Myo16 in the nucleus remain unknown. In this report, we investigated Myo16 abundance during transit through the cell cycle. Immunolocalization, immunoblot, flow cytometric and quantitative RT-PCR studies performed in Rat2 cells indicate that Myo16 mRNA and protein abundance are cell cycle regulated: in the unperturbed cell cycle, each rises to peak levels in late G1 and thereon through S-phase and each decays as cells enter M-phase. Notably, RNA interference-induced Myo16 depletion results in altered cell cycle distribution as well as in large-scale cell death. In response to DNA replication stress (impaired replication fork progression as a consequence of DNA damage, lack of sufficient deoxynucleotides, or inhibition of DNA polymerases), Myo16 protein shows substantial loss. Attenuation of replication stress (aphidicolin or hydroxyurea) is followed by a recovery of Myo16 expression and resumption of S-phase progression. Collectively, these observations suggest that Myo16 may play a regulatory role in cell cycle progression.
Collapse
Affiliation(s)
- Richard S Cameron
- Institute of Molecular Medicine and Genetics, Department of Medicine, Georgia Regents University, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | | | |
Collapse
|
36
|
Wiedner SD, Vedejs E. Reactivity of aziridinomitosene derivatives related to FK317 in the presence of protic nucleophiles. J Org Chem 2012; 77:1045-55. [PMID: 22208619 PMCID: PMC3264803 DOI: 10.1021/jo202286a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The syntheses and reactivity of N-TBDPS and N-trityl protected derivatives of an aziridinomitosene corresponding to FK317 are described. New reactivity patterns were observed for these highly sensitive and functionally dense heterocycles under mild nucleophilic conditions approaching the threshold for degradation. Thus, the silyl or trityl protected aziridinomitosene reacted with Cs(2)CO(3)/CD(3)OD to give isomeric products where substitution occurred at C(10) and C(9a) (mitomycin numbering) providing a CD(3) ether and a CD(3) hemiaminal, respectively. These findings show that heterolysis at C(10) is faster than at aziridine C(1), in contrast to the behavior of typical aziridinomitosenes in the mitomycin series. The labile N-TBDPS hemiaminal and the more stable N-trityl hemiaminal resemble the mitomycin K substitution pattern. A reagent consisting of CsF in CF(3)CH(2)OH/CH(3)CN desilylated a simple N-TBDPS aziridine but caused nucleophilic cleavage at C(1) as well as C(10) without cleavage of the N-TBPDS group in the fully functionalized penultimate aziridinomitosene. The high reactivity of the C(10) carbamate with nucleophiles precludes the use of deprotection methodology that requires N-protonation for fully functionalized aziridinomitosenes in the FK317 series.
Collapse
Affiliation(s)
| | - Edwin Vedejs
- Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109
| |
Collapse
|
37
|
Paz MM, Pritsos CA. The Molecular Toxicology of Mitomycin C. ADVANCES IN MOLECULAR TOXICOLOGY VOLUME 6 2012. [DOI: 10.1016/b978-0-444-59389-4.00007-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|