1
|
Siutkina A, Kalinina S, Liu R, Heitman LH, Junker A, Daniliuc CG, Kalinin DV. Microwave-Assisted Synthesis, Structure, and Preliminary Biological Evaluation of Novel 6-Methoxy-5,6-dihydro-5-azapurines. ACS OMEGA 2023; 8:14097-14112. [PMID: 37091407 PMCID: PMC10116508 DOI: 10.1021/acsomega.3c00765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/10/2023] [Indexed: 05/03/2023]
Abstract
We herein disclose the microwave-assisted synthesis of previously unreported 6-methoxy-5,6-dihydro-5-azapurines, whose purine-like scaffold is promising for drug discovery. The method is simple, fast, and relies on easily accessible reagents such as trimethyl orthoformate, acetic acid, and aminotriazole-derived N,N'-disubstituted formamidines. The preliminary biological evaluation revealed that selected representatives of synthesized 6-methoxy-5,6-dihydro-5-azapurines dose-dependently reduce the viability of HepG2 and A549 cancer cells having little to no influence on five tested purinergic receptors.
Collapse
Affiliation(s)
- Alena
I. Siutkina
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany
| | - Svetlana Kalinina
- Institute
of Food Chemistry, University of Münster, 48149 Münster, Germany
| | - Rongfang Liu
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 CC Leiden, The Netherlands
| | - Laura H. Heitman
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 CC Leiden, The Netherlands
| | - Anna Junker
- European
Institute for Molecular Imaging (EIMI), University of Münster, 48149 Münster, Germany
| | | | - Dmitrii V. Kalinin
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany
| |
Collapse
|
2
|
Wang Y, Katanski CD, Watkins C, Pan JN, Dai Q, Jiang Z, Pan T. A high-throughput screening method for evolving a demethylase enzyme with improved and new functionalities. Nucleic Acids Res 2021; 49:e30. [PMID: 33337498 PMCID: PMC7968990 DOI: 10.1093/nar/gkaa1213] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 01/26/2023] Open
Abstract
AlkB is a DNA/RNA repair enzyme that removes base alkylations such as N1-methyladenosine (m1A) or N3-methylcytosine (m3C) from DNA and RNA. The AlkB enzyme has been used as a critical tool to facilitate tRNA sequencing and identification of mRNA modifications. As a tool, AlkB mutants with better reactivity and new functionalities are highly desired; however, previous identification of such AlkB mutants was based on the classical approach of targeted mutagenesis. Here, we introduce a high-throughput screening method to evaluate libraries of AlkB variants for demethylation activity on RNA and DNA substrates. This method is based on a fluorogenic RNA aptamer with an internal modified RNA/DNA residue which can block reverse transcription or introduce mutations leading to loss of fluorescence inherent in the cDNA product. Demethylation by an AlkB variant eliminates the blockage or mutation thereby restores the fluorescence signals. We applied our screening method to sites D135 and R210 in the Escherichia coli AlkB protein and identified a variant with improved activity beyond a previously known hyperactive mutant toward N1-methylguanosine (m1G) in RNA. We also applied our method to O6-methylguanosine (O6mG) modified DNA substrates and identified candidate AlkB variants with demethylating activity. Our study provides a high-throughput screening method for in vitro evolution of any demethylase enzyme.
Collapse
Affiliation(s)
- Yuru Wang
- Department of Biochemistry and Molecular Biology, USA.,Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | | | | | - Jessica N Pan
- Department of Biochemistry and Molecular Biology, USA
| | - Qing Dai
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Zhuoxun Jiang
- Department of Biochemistry and Molecular Biology, USA
| | - Tao Pan
- Department of Biochemistry and Molecular Biology, USA
| |
Collapse
|
3
|
Grafström RC, Edman CC, Sundqvist K, Liu Y, Hybbinette SS, Atzori L, Nicotera P, Dypbukt JM. Cultured Human Bronchial Cells as a Model System in Lung Toxicology and Carcinogenesis: Implications from Studies with Acrolein. Altern Lab Anim 2020. [DOI: 10.1177/026119298901600305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Tissues and cells from both humans and laboratory animals can now be successfully cultured and used to examine in vitro the interspecial extrapolation of toxicity data. In particular, epithelial cells or fibroblasts from the human bronchus are systematically being used to study pathobiological effects of inhalation toxicants, e.g. acrolein, present in tobacco smoke and automobile emissions. Micromolar concentrations of acrolein are highly toxic to both bronchial epithelial cells and fibroblasts, and decrease their colony-forming efficiency, ability to exclude trypan blue dye, and content of low-molecular-weight thiols in a dose-dependent manner. Cytosolic free-calcium levels are increased at concentrations of acrolein lower than those required to decrease plasma membrane integrity (measured by exclusion of ethidium bromide dye). Acrolein also decreases growth rate and increases formation of cross-linked envelopes in bronchial epithelial cells, which are indicative of squamous differentiation. Moreover, acrolein causes several types of DNA damage, including single strand breaks, DNA-protein cross-links, interstrand cross-links and alkali-labile sites. The dose-response relationships in this study indicate that disturbance of the cellular thiol and calcium homeostasis and/or formation of DNA damage may be involved in acrolein-related toxicity and accelerated epithelial differentiation. These results also demonstrate that acrolein causes pathobiological effects associated with various phases of multi-stage carcinogenesis in human airway epithelium.
Collapse
Affiliation(s)
- Roland C. Grafström
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Charlotte C. Edman
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Kristina Sundqvist
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Yun Liu
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Sverker S. Hybbinette
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Luigi Atzori
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Pierluigi Nicotera
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| | - Jeannette M. Dypbukt
- Department of Toxicology, Karolinska Institute, P.O. Box 60400, S-104 01 Stockholm, Sweden
| |
Collapse
|
4
|
Lee HT, Sanford S, Paul T, Choe J, Bose A, Opresko PL, Myong S. Position-Dependent Effect of Guanine Base Damage and Mutations on Telomeric G-Quadruplex and Telomerase Extension. Biochemistry 2020; 59:2627-2639. [PMID: 32578995 DOI: 10.1021/acs.biochem.0c00434] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Telomeres are hot spots for mutagenic oxidative and methylation base damage due to their high guanine content. We used single-molecule fluorescence resonance energy transfer detection and biochemical assays to determine how different positions and types of guanine damage and mutations alter telomeric G-quadruplex structure and telomerase activity. We compared 15 modifications, including 8-oxoguanine (8oxoG), O-6-methylguanine (O6mG), and all three possible point mutations (G to A, T, and C) at the 3' three terminal guanine positions of a telomeric G-quadruplex, which is the critical access point for telomerase. We found that G-quadruplex structural instability was induced in the order C < T < A ≤ 8oxoG < O6mG, with the perturbation caused by O6mG far exceeding the perturbation caused by other base alterations. For all base modifications, the central G position was the most destabilizing among the three terminal guanines. While the structural disruption by 8oxoG and O6mG led to concomitant increases in telomerase binding and extension activity, the structural perturbation by point mutations (A, T, and C) did not, due to disrupted annealing between the telomeric overhang and the telomerase RNA template. Repositioning the same mutations away from the terminal guanines caused both G-quadruplex structural instability and elevated telomerase activity. Our findings demonstrate how a single-base modification drives structural alterations and telomere lengthening in a position-dependent manner. Furthermore, our results suggest a long-term and inheritable effect of telomeric DNA damage that can lead to telomere lengthening, which potentially contributes to oncogenesis.
Collapse
Affiliation(s)
- Hui-Ting Lee
- Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Samantha Sanford
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health and University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania 15261, United States
| | - Tapas Paul
- Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Joshua Choe
- Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Arindam Bose
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health and University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania 15261, United States
| | - Patricia L Opresko
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health and University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania 15261, United States
| | - Sua Myong
- Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Physics Frontier Center (Center for Physics of Living Cells), University of Illinois, 1110 West Green Street, Urbana, Illinois 61801, United States
| |
Collapse
|
5
|
Stachelska-Wierzchowska A, Wierzchowski J, Górka M, Bzowska A, Wielgus-Kutrowska B. Tri-Cyclic Nucleobase Analogs and their Ribosides as Substrates of Purine-Nucleoside Phosphorylases. II Guanine and Isoguanine Derivatives. Molecules 2019; 24:E1493. [PMID: 30995785 PMCID: PMC6514686 DOI: 10.3390/molecules24081493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/27/2019] [Accepted: 04/09/2019] [Indexed: 11/17/2022] Open
Abstract
Etheno-derivatives of guanine, O6-methylguanine, and isoguanine were prepared and purified using standard methods. The title compounds were examined as potential substrates of purine-nucleoside phosphorylases from various sources in the reverse (synthetic) pathway. It was found that 1,N2-etheno-guanine and 1,N6-etheno-isoguanine are excellent substrates for purine-nucleoside phosphorylase (PNP) from E. coli, while O6-methyl-N2,3-etheno-guanine exhibited moderate activity vs. this enzyme. The latter two compounds displayed intense fluorescence in neutral aqueous medium, and so did the corresponding ribosylation products. By contrast, PNP from calf spleens exhibited only modest activity towards 1,N6-etheno-isoguanine; the remaining compounds were not ribosylated by this enzyme. The enzymatic ribosylation of 1,N6-etheno-isoguanine using two forms of calf PNP (wild type and N243D) and E. coli PNP (wild type and D204N) gave three different products, which were identified on the basis of NMR analysis and comparison with the product of the isoguanosine reaction with chloroacetic aldehyde, which gave an essentially single compound, identified unequivocally as N9-riboside. With the wild-type E. coli enzyme as a catalyst, N9--d- and N7--d-ribosides are obtained in proportion ~1:3, while calf PNP produced another riboside, tentatively identified as N6--d-riboside. The potential application of various forms of PNP for synthesis of the tri-cyclic nucleoside analogs is discussed.
Collapse
Affiliation(s)
- Alicja Stachelska-Wierzchowska
- Department of Physics and Biophysics, University of Varmia & Masuria in Olsztyn, 4 Oczapowskiego St., 10-719 Olsztyn, Poland.
| | - Jacek Wierzchowski
- Department of Physics and Biophysics, University of Varmia & Masuria in Olsztyn, 4 Oczapowskiego St., 10-719 Olsztyn, Poland.
| | - Michał Górka
- Division of Biophysics, Institute of Experimental Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland.
- Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warsaw, Poland.
| | - Agnieszka Bzowska
- Division of Biophysics, Institute of Experimental Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland.
| | - Beata Wielgus-Kutrowska
- Division of Biophysics, Institute of Experimental Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland.
| |
Collapse
|
6
|
Matsuno A, Murakami M, Hoya K, Yamada SM, Miyamoto S, Yamada S, Son JH, Nishido H, Ide F, Nagashima H, Sugaya M, Hirohata T, Mizutani A, Okinaga H, Ishii Y, Tahara S, Teramoto A, Osamura RY. Molecular status of pituitary carcinoma and atypical adenoma that contributes the effectiveness of temozolomide. Med Mol Morphol 2013; 47:1-7. [PMID: 23955641 DOI: 10.1007/s00795-013-0050-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/02/2013] [Indexed: 01/16/2023]
Abstract
There have been several reports of temozolomide (TMZ) treatment of pituitary carcinomas and atypical adenomas. O(6)-methyl-guanine-DNA methyltransferase is not the sole molecule determining the sensitivity to TMZ in pituitary carcinomas and atypical adenomas. The Japan Society of Hypothalamic and Pituitary Tumors study suggests that MSH6, one of mismatch repair pathway enzyme, fulfills a contributory role to the efficacy of TMZ treatment for pituitary carcinomas and atypical adenomas. The preserved MSH6 function might be essential for the responsiveness to TMZ treatment in pituitary carcinomas and atypical adenomas.
Collapse
Affiliation(s)
- Akira Matsuno
- Department of Neurosurgery, Teikyo University Chiba Medical Center, 3426-3 Anesaki, Ichihara, Chiba, 299-0111, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Iglesias E, Casado J. Mechanisms of hydrolysis and nitrosation reactions of alkyl nitrites in various media. INT REV PHYS CHEM 2010. [DOI: 10.1080/01442350110092693] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
8
|
Reithmeier T, Graf E, Piroth T, Trippel M, Pinsker MO, Nikkhah G. BCNU for recurrent glioblastoma multiforme: efficacy, toxicity and prognostic factors. BMC Cancer 2010; 10:30. [PMID: 20122270 PMCID: PMC2837009 DOI: 10.1186/1471-2407-10-30] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2009] [Accepted: 02/02/2010] [Indexed: 11/23/2022] Open
Abstract
Background The prognosis for patients with recurrent glioblastoma is still poor with a median survival between 3 and 6 months. Reports about the application of carmustine (BCNU), one of the standard chemotherapeutic drugs in the treatment of newly diagnosed glioblastoma, in the recurrent situation are rare. Methods We performed a retrospective analysis of 35 patients with recurrent or progressive glioblastoma treated with 80 mg/m2 BCNU on days 1 on 3 intravenously at our department for efficacy, toxicity and prognostic factors. Progression free survival and overall survival were estimated by the Kaplan-Meier method. The influence of age, Karnofsky performance status (KPS), tumor burden, pretreatment with temozolomide (TMZ), type of surgery for initial diagnosis and number of previous relapses on outcome was analyzed in a proportional hazards regression model. Results The median age of the group was 53 years, median KPS was 70. Median progression free survival was 11 weeks (95% confidence interval [CI]: 8-15), median overall survival 22 weeks (95% CI: 18-27). The rate of adverse events, especially hematological toxicity, is relatively high, and in 3 patients treatment had to be terminated due to adverse events (one pulmonary embolism, one pulmonary fibrosis, and one severe bone marrow suppression). No influence of age, KPS, tumor burden, pre-treatment with TMZ and number of previous relapses on outcome could be demonstrated, while gross total resection prior to recurrence showed a borderline statistically significant negative impact on PFS and OS. These data compare well with historical survival figures. However prospective randomized studies are needed to evaluate BCNU efficacy against newer drugs like bevacizumab or the intensified temozolomide regime (one week on/one week off). Conclusion In summary, BCNU treatment appears to be a valuable therapeutic option for recurrent glioblastomas, where no other validated radio- and/or chemotherapy are available.
Collapse
Affiliation(s)
- Thomas Reithmeier
- Department of Stereotactic and Functional Neurosurgery, University Medical Center Freiburg, Breisacher Str, 64, 79106 Freiburg im Breisgau, Germany.
| | | | | | | | | | | |
Collapse
|
9
|
Lorente A, Mueller W, Urdangarín E, Lázcoz P, Lass U, von Deimling A, Castresana JS. RASSF1A, BLU, NORE1A, PTEN and MGMT expression and promoter methylation in gliomas and glioma cell lines and evidence of deregulated expression of de novo DNMTs. Brain Pathol 2008; 19:279-92. [PMID: 18616639 DOI: 10.1111/j.1750-3639.2008.00185.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Methylation of CpG islands in gene promoters can lead to gene silencing. Together with deletion or mutation, it may cause a loss of function of tumor suppressor genes. RASSF1A (3p21.3), NORE1A (1q32.1) and BLU (3p21.3) have been shown to be downregulated by methylation in cancer, and PTEN (10q23.3) and MGMT (10q26.1) are located in areas commonly deleted in astrocytomas. MGMT methylation predicts a better response and a longer overall survival in patients with glioblastomas treated with temozolomide. We analyzed 53 astrocytoma samples and 10 high-grade glioma cell lines. Gene expression was assessed by RT-PCR. Bisulfite sequencing, MSP and a melting curve analysis-based real-time PCR were performed to detect promoter methylation. Treatments with 5'-aza-2'-deoxicitidine were applied to restore gene expression in cell lines. Ninety-two percent of tumor samples were methylated for RASSF1A, 30%-57% for BLU and 47% for MGMT, suggesting promoter methylation of these genes to be a common event in glioma tumorigenesis. Only 4% of the tumors revealed a methylated promoter for NORE1A. No association between methylation and loss of expression could be established for PTEN. We identified de novo DNMTs overexpression in a subset of tumors which may explain the methylation phenotype of individual gliomas.
Collapse
Affiliation(s)
- Aiala Lorente
- Brain Tumor Biology Unit, University of Navarra, Irunlarrea 1, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
10
|
Russell LB, Hunsicker PR, Russell WL. Comparison of the genetic effects of equimolar doses of ENU and MNU: while the chemicals differ dramatically in their mutagenicity in stem-cell spermatogonia, both elicit very high mutation rates in differentiating spermatogonia. Mutat Res 2007; 616:181-95. [PMID: 17174358 PMCID: PMC1905495 DOI: 10.1016/j.mrfmmm.2006.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Mutagenic, reproductive, and toxicity effects of two closely related chemicals, ethylnitrosourea (ENU) and methylnitrosourea (MNU), were compared at equimolar and near-equimolar doses in the mouse specific-locus test in a screen of all stages of spermatogenesis and spermiogenesis. In stem-cell spermatogonia (SG), ENU is more than an order of magnitude more mutagenic than MNU. During post-SG stages, both chemicals exhibit high peaks in mutation yield when differentiating spermatogonia (DG) and preleptotene spermatocytes are exposed. The mutation frequency induced by 75mgMNU/kg during this peak interval is, to date, the highest induced by any single-exposure mutagenic treatment - chemical or radiation - that allows survival of the exposed animal and its germ cells, producing an estimated 10 new mutations per genome. There is thus a vast difference between stem cell and differentiating spermatogonia in their sensitivity to MNU, but little difference between these stages in their sensitivity to ENU. During stages following meiotic metaphase, the highest mutation yield is obtained from exposed spermatids, but for both chemicals, that yield is less than one-quarter that obtained from the peak interval. Large-lesion (LL) mutations were induced only in spermatids. Although only a few of the remaining mutations were analyzed molecularly, there is considerable evidence from recent molecular characterizations of the marker genes and their flanking chromosomal regions that most, if not all, mutations induced during the peak-sensitive period did not involve lesions outside the marked loci. Both ENU and MNU treatments of post-SG stages yielded significant numbers of mutants that were recovered as mosaics, with the proportion being higher for ENU than for MNU. Comparing the chemicals for the endpoints studied and additional ones (e.g., chromosome aberrations, toxicity to germ cells and to animals, teratogenicity) revealed that while MNU is generally more effective, the opposite is true when the target cells are SG.
Collapse
Affiliation(s)
- Liane B Russell
- Life Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831-6420, USA.
| | | | | |
Collapse
|
11
|
Preusser M, Haberler C, Hainfellner JA. Malignant glioma: Neuropathology and Neurobiology. Wien Med Wochenschr 2006; 156:332-7. [PMID: 16944363 DOI: 10.1007/s10354-006-0304-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Accepted: 03/31/2006] [Indexed: 12/17/2022]
Abstract
Malignant gliomas may manifest at any age including congenital and childhood cases. Peak incidence is, however, in adults older than 40 years. Males are more frequently affected than females. The sole unequivocal risk factor is therapeutic ionizing irradiation. Malignant gliomas comprise a spectrum of different tumor subtypes. Within this spectrum, glioblastoma, anaplastic astrocytoma and anaplastic oligodendroglioma share as basic features preferential location in cerebral hemispheres, diffuse infiltration of brain tissue, fast tumor growth with fatal outcome within months or years. Invasion is regarded as one of the main reasons for poor therapeutic success, because it makes complete surgical removal of gliomas impossible. Invasion of glioma cells requires interaction with the extracellular matrix and with surrounding cells of the healthy brain tissue. Vascular proliferates and tissue necrosis are characteristic features of malignant gliomas, in particular glioblastoma. These features are most likely the consequence of rapidly increasing tumor mass that is inadequately oxygenized by the preexisting vasculature. In malignant glioma, distinct molecular pathways including the p53 pathway, the RB pathway and the EGFR pathway show frequent alterations that seem to be pathogenetically relevant. Methylguanine-methyltransferase (MGMT) promoter methylation status in glioblastoma and 1p19q deletion status in anaplastic oligodendroglioma are associated with response to chemotherapy. The role of neuropathology and neurobiology in neurooncology is 1. to provide a clinically meaningful classification of brain tumors on basis of pathobiological factors, 2. to clarify etiology and pathogenesis of brain tumors as rational basis for development of new diagnostic tests and therapies, and 3. to translate testing for new clinically relevant molecular parameters into clinical application.
Collapse
Affiliation(s)
- Matthias Preusser
- Institute of Neurology, Medical University Vienna, Währinger Gürtel 18-20, 1097 Vienna, Austria
| | | | | |
Collapse
|
12
|
Affiliation(s)
- Yukiko Mishina
- Department of Chemistry, The University of Chicago, 5735 South Ellis Avenue, Chicago, Illinois 60637
| | - Erica M. Duguid
- Department of Chemistry, The University of Chicago, 5735 South Ellis Avenue, Chicago, Illinois 60637
| | - Chuan He
- Department of Chemistry, The University of Chicago, 5735 South Ellis Avenue, Chicago, Illinois 60637
| |
Collapse
|
13
|
Abstract
O(6)-Methylguanine DNA methyltransferase (MGMT) has been studied for >20 years as a gene that is associated with the mutagenicity and cytotoxicity induced by either methylating carcinogens or alkylating (methylating and chloroethylating) therapeutic agents. Pioneering studies of alkylating agents identified alkylated guanine at the O(6) position, the substrate of MGMT, as a potentially promutagenic and lethal toxic DNA lesion. MGMT plays a prominent role in DNA adduct repair that limits the mutagenic and cytotoxic effect of alkylating agents. Because of its role in cancer etiology and chemotherapy resistance, MGMT is of particular interest. In this article, the clinical effect of MGMT expression and targeted modulation of MGMT will be summarized.
Collapse
Affiliation(s)
- Lili Liu
- Department of Medicine, Division of Hematology/Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
14
|
Dimopoulos MA, Souliotis VL, Anagnostopoulos A, Papadimitriou C, Sfikakis PP. Extent of damage and repair in the p53 tumor-suppressor gene after treatment of myeloma patients with high-dose melphalan and autologous blood stem-cell transplantation is individualized and may predict clinical outcome. J Clin Oncol 2005; 23:4381-9. [PMID: 15883412 DOI: 10.1200/jco.2005.07.385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To quantitate the individual levels of melphalan-induced DNA damage formation and repair in vivo and to search for possible correlations with clinical outcome in patients with multiple myeloma (MM). PATIENTS AND METHODS The formation and subsequent repair of DNA damage (monoadducts and interstrand cross-links) in the p53 tumor-suppressor gene, the proto-oncogene N-ras, and the housekeeping gene beta-actin during the first 24 hours after treatment with high-dose melphalan (HDM; 200 mg/m2) supported by autologous blood stem-cell transplantation (ABSCT) was measured in blood leukocytes of 26 patients with MM. The peak DNA adduct levels, the total amount of adducts over time, and the rate of adducts repair in each gene were correlated with response and time to progression after HDM. RESULTS The levels of gene-specific DNA damage formation and the individual repairing capacity varied up to 16-fold among patients, indicating that the melphalan-induced biologic effect in vivo is highly individualized. A significantly greater DNA damage and a slower rate of repair in p53 for all end points under study were found in patients who achieved tumor reduction compared with nonresponding patients. Furthermore, longer progression-free survival correlated with increased peak monoadduct levels in the p53 gene (P = .032). CONCLUSION Increased DNA damage and slower repairing capacity in the p53 gene from blood leukocytes after HDM correlate with improved outcome of patients with MM who undergo ABSCT. These results suggest that quantitation of such biologic end points may identify patients who are more likely to benefit from this procedure.
Collapse
Affiliation(s)
- Meletios A Dimopoulos
- Department of Clinical Therapeutics, University of Athens School of Medicine, Greece.
| | | | | | | | | |
Collapse
|
15
|
García‐Río L, Hervés P, Leis JR, Mejuto JC, Rodríguez‐Dafonte P. Reactive micelles: nitroso group transfer from
N
‐methyl‐
N
‐nitroso‐
p
‐toluenesulfonamide to amphiphilic amines. J PHYS ORG CHEM 2004. [DOI: 10.1002/poc.827] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- L. García‐Río
- Departamento de Química Física, Facultad de Química, Universidad de Santiago de Compostela, 15706‐Santiago de Compostela, Spain
| | - P. Hervés
- Departamento de Química Física, Facultad de Ciencias, Universidad de Vigo, 32004‐Ourense, Spain
| | - J. R. Leis
- Departamento de Química Física, Facultad de Química, Universidad de Santiago de Compostela, 15706‐Santiago de Compostela, Spain
| | - J. C. Mejuto
- Departamento de Química Física, Facultad de Ciencias, Universidad de Vigo, 32004‐Ourense, Spain
| | - P. Rodríguez‐Dafonte
- Departamento de Química Física, Facultad de Química, Universidad de Santiago de Compostela, 15706‐Santiago de Compostela, Spain
| |
Collapse
|
16
|
O
6
-Methylguanine-DNA Methyltransferase Gene: Epigenetic Silencing and Prognostic Value in Head and Neck Squamous Cell Carcinoma. Cancer Epidemiol Biomarkers Prev 2004. [DOI: 10.1158/1055-9965.967.13.6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background: Alkylating N-nitroso compounds can interact directly with DNA, forming O6-alkylguanine, a DNA adduct proved to be mutagenic and carcinogenic if not sufficiently repaired. A specific DNA repair enzyme, O6-methylguanine-DNA methyltransferase (MGMT), can remove the alkyl group from the O6-position of the guanine, thereby preventing its mutagenic and carcinogenic effects. Inactivation of the MGMT gene in association with promoter hypermethylation results in persistence of O6-alkylguanine in DNA, leading to G:C to A:T transition mutation and these G:C to A:T transition mutations can inactivate p53 tumor suppressor gene or activate ras proto-oncogene. Methods: We analyzed MGMT promoter hypermethylation and protein expression patterns in 94 cases of primary head and neck squamous cell carcinoma (HNSCC) by methylation-specific PCR (MSP) and immunohistochemical staining. The results were then correlated with clinical follow-up data. Results: MGMT promoter hypermethylation was present in 17 of 94 patients (18.1%) and apparent loss of protein expression was seen in 19 of 93 HNSCC patients (20.4%). The presence of MGMT promoter hypermethylation was significantly correlated with loss of MGMT protein expression in HNSCC. Both MGMT promoter hypermethylation and loss of protein expression were significantly correlated to increased tumor recurrences and decreased patient survival, independent of other risk factors, such as tumor site, tumor size, nodal status, age, and chemoradiation therapy. Conclusions: MGMT promoter hypermethylation and apparent loss of protein expression are reliable and independent prognostic factors in HNSCC. The above study may also provide guideline or basis for applying alkylating antitumor agents to patients with HNSCC that display MGMT promoter hypermethylation and/or loss of MGMT protein expression.
Collapse
|
17
|
Park TJ, Paik WK, Lim IK. Induction of thermal and chemical stability of O6-methylguanine-DNA methyltransferase by Ca2+. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1599:36-44. [PMID: 12479403 DOI: 10.1016/s1570-9639(02)00379-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Divalent cations stabilized rat recombinant O6-methylguanine-DNA methyltransferase (rMGMT) protein against heat treatment. Activity of rMGMT was completely abolished by incubating at 45 degrees C for 30 min, however, addition of 1.0 mM Mg2+, Ca2+ or Mn2+ significantly protected heat-induced inactivation of MGMT activity (50-60% vs. 97% inactivation). Protective effect of Ca2+ on the MGMT activity was concentration-dependent up to 3 mM, and the thermal protection was effective up to 45 degrees C. In order to investigate Ca2+ binding site in rMGMT protein, truncated GST-rMGMT proteins containing N-terminal 39 amino acids (GST-rMGMT39), 70 amino acids (GST-rMGMT70) and full-length protein (GST-rMGMT) were prepared. Radiolabeled calcium ion [45Ca2+] was bound only to the GST-rMGMT70 and GST-rMGMT, but not to the GST-rMGMT39, indicating that divalent cations could bind the residues between 40th and 70th of the rMGMT protein. Calcium binding was not observed in the site-directed mutant rMGMT proteins (rMGMT(D42A) and rMGMT(E45A)), confirmed by autoradiography using [45Ca2+] after nondenaturing gel electrophoresis; however, the above two mutants had the same catalytic activity as well as proteolytic sensitivity as the wild MGMT protein. Analysis by equilibrium dialysis revealed stoichiometric binding of one molecule of Ca2+ to one molecule of the protein. Since circular dichroism (CD) spectra indicated no discernible difference before and after Ca2+ binding, the above results suggested that neutralization of two negative charges of Asp42 and Glu45 by Ca2+ resulted in thermal stabilization of the protein with minimum perturbation of its tertiary structure.
Collapse
Affiliation(s)
- Tae Jun Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 442-721, Gyunggi-do, South Korea
| | | | | |
Collapse
|
18
|
Quinn JA, Pluda J, Dolan ME, Delaney S, Kaplan R, Rich JN, Friedman AH, Reardon DA, Sampson JH, Colvin OM, Haglund MM, Pegg AE, Moschel RC, McLendon RE, Provenzale JM, Gururangan S, Tourt-Uhlig S, Herndon JE, Bigner DD, Friedman HS. Phase II trial of carmustine plus O(6)-benzylguanine for patients with nitrosourea-resistant recurrent or progressive malignant glioma. J Clin Oncol 2002; 20:2277-83. [PMID: 11980998 DOI: 10.1200/jco.2002.09.084] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE We conducted a phase II trial of carmustine (BCNU) plus the O(6)-alkylguanine-DNA alkyltransferase inhibitor O(6)-benzylguanine (O(6)-BG) to define the activity and toxicity of this regimen in the treatment of adults with progressive or recurrent malignant glioma resistant to nitrosoureas. PATIENTS AND METHODS Patients were treated with O(6)-BG at an intravenous dose of 120 mg/m(2) followed 1 hour later by 40 mg/m(2) of BCNU, with cycles repeated at 6-week intervals. RESULTS Eighteen patients were treated (15 with glioblastoma multiforme, two with anaplastic astrocytoma, and one with malignant glioma). None of the 18 patients demonstrated a partial or complete response. Two patients exhibited stable disease for 12 weeks before their tumors progressed. Three patients demonstrated stable disease for 6, 12, and 18 weeks before discontinuing therapy because of hematopoietic toxicity. Twelve patients experienced reversible > or = grade 3 hematopoietic toxicity. There was no difference in half-lives (0.56 +/- 0.21 hour v 0.54 +/- 0.20 hour) or area under the curve values (4.8 +/- 1.7 microg/mL/h v 5.0 +/- 1.3 microg/mL/h) of O(6)-BG for patients receiving phenytoin and those not treated with this drug. CONCLUSION These results indicate that O(6)-BG plus BCNU at the dose schedule used in this trial is unsuccessful in producing tumor regression in patients with nitrosourea-resistant malignant glioma, although stable disease was seen in five patients for 6, 12, 12, 12, and 18 weeks. Future use of this approach will require strategies to minimize dose-limiting toxicity of BCNU such as regional delivery or hematopoietic stem-cell protection.
Collapse
Affiliation(s)
- Jennifer A Quinn
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhou ZQ, Manguino D, Kewitt K, Intano GW, McMahan CA, Herbert DC, Hanes M, Reddick R, Ikeno Y, Walter CA. Spontaneous hepatocellular carcinoma is reduced in transgenic mice overexpressing human O6- methylguanine-DNA methyltransferase. Proc Natl Acad Sci U S A 2001; 98:12566-71. [PMID: 11606727 PMCID: PMC60094 DOI: 10.1073/pnas.221232998] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
O(6)-methylguanine (O(6)mG) is a potent mutagenic and procarcinogenic DNA lesion. Organisms have evolved with a DNA repair mechanism that largely ameliorates the deleterious effects of O(6)mG through a direct reversal mechanism by a protein termed O(6)-methylguanine-DNA methyltransferase (MGMT). However, the contribution of O(6)mG to carcinogenesis, in the absence of known exposure to agents that produce it, has not been defined. Nontransgenic C3HeB male mice have a high frequency of spontaneous liver tumors. Transgenic CeHeB/FeJ mice expressing human MGMT (hMGMT) were generated that had elevated hepatic MGMT activity. The spontaneous development of hepatocellular carcinoma was significantly reduced in those mice expressing hMGMT compared with nontransgenic C3HeB/FeJ male mice. No differences were detected in spontaneous mutant frequencies in lacI transgenes in mice carrying hMGMT compared with that without hMGMT but the proportion of GC to AT transition mutations was lower in the transgenic mice carrying hMGMT as well as lacI. Tumors that arose in C3HeB/FeJ transgenic mice were largely deficient in hMGMT protein as determined by immunohistochemistry with a monoclonal antibody directed against hMGMT. Together these data indicate that spontaneous O(6)mG lesions induced hepatocellular carcinogenesis in C3HeB/FeJ male mice. These transgenic mice represent a rare example of reduced spontaneous carcinogenesis.
Collapse
Affiliation(s)
- Z Q Zhou
- Department of Cellular and Structural Biology, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Cells are constantly under threat from the cytotoxic and mutagenic effects of DNA damaging agents. These agents can either be exogenous or formed within cells. Environmental DNA-damaging agents include UV light and ionizing radiation, as well as a variety of chemicals encountered in foodstuffs, or as air- and water-borne agents. Endogenous damaging agents include methylating species and the reactive oxygen species that arise during respiration. Although diverse responses are elicited in cells following DNA damage, this review focuses on three aspects: DNA repair mechanisms, cell cycle checkpoints, and apoptosis. Because the areas of nucleotide excision repair and mismatch repair have been covered extensively in recent reviews, we restrict our coverage of the DNA repair field to base excision repair and DNA double-strand break repair.
Collapse
Affiliation(s)
- C J Norbury
- Imperial Cancer Research Fund Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, United Kingdom.
| | | |
Collapse
|
21
|
Christmann M, Pick M, Lage H, Schadendorf D, Bernd Kaina. Acquired resistance of melanoma cells to the antineoplastic agent fotemustine is caused by reactivation of the DNA repair gene mgmt. Int J Cancer 2001. [DOI: 10.1002/1097-0215(200102)9999:9999<::aid-ijc1160>3.0.co;2-v] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Friedman HS, Pluda J, Quinn JA, Ewesuedo RB, Long L, Friedman AH, Cokgor I, Colvin OM, Haglund MM, Ashley DM, Rich JN, Sampson J, Pegg AE, Moschel RC, McLendon RE, Provenzale JM, Stewart ES, Tourt-Uhlig S, Garcia-Turner AM, Herndon JE, Bigner DD, Dolan ME. Phase I trial of carmustine plus O6-benzylguanine for patients with recurrent or progressive malignant glioma. J Clin Oncol 2000; 18:3522-8. [PMID: 11032594 DOI: 10.1200/jco.2000.18.20.3522] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The major mechanism of resistance to alkylnitrosourea therapy involves the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT), which removes chloroethylation or methylation damage from the O(6) position of guanine. O(6)-benzylguanine (O(6)-BG) is an AGT substrate that inhibits AGT by suicide inactivation. We conducted a phase I trial of carmustine (BCNU) plus O(6)-BG to define the toxicity and maximum-tolerated dose (MTD) of BCNU in conjunction with the preadministration of O(6)-BG with recurrent or progressive malignant glioma. PATIENTS AND METHODS Patients were treated with O(6)-BG at a dose of 100 mg/m(2) followed 1 hour later by BCNU. Cohorts of three to six patients were treated with escalating doses of BCNU, and patients were observed for at least 6 weeks before being considered assessable for toxicity. Plasma samples were collected and analyzed for O(6)-BG, 8-oxo-O(6)-BG, and 8-oxoguanine concentration. RESULTS Twenty-three patients were treated (22 with glioblastoma multiforme and one with anaplastic astrocytoma). Four dose levels of BCNU (13.5, 27, 40, and 55 mg/m(2)) were evaluated, with the highest dose level being complicated by grade 3 or 4 thrombocytopenia and neutropenia. O(6)-BG rapidly disappeared from plasma (elimination half-life = 0. 54 +/- 0.14 hours) and was converted to a longer-lived metabolite, 8-oxo-O(6)-BG (elimination half-life = 5.6 +/- 2.7 hours) and further to 8-oxoguanine. There was no detectable O(6)-BG 5 hours after the start of the O(6)-BG infusion; however, 8-oxo-O(6)-BG and 8-oxoguanine concentrations were detected 25 hours after O(6)-BG infusion. The mean area under the concentration-time curve (AUC) of 8-oxo-O(6)-BG was 17.5 times greater than the mean AUC for O(6)-BG. CONCLUSION These results indicate that the MTD of BCNU when given in combination with O(6)-BG at a dose of 100 mg/m(2) is 40 mg/m(2) administered at 6-week intervals. This study provides the foundation for a phase II trial of O(6)-BG plus BCNU in nitrosourea-resistant malignant glioma.
Collapse
Affiliation(s)
- H S Friedman
- Departments of Surgery, Medicine, Pathology, Radiology, and Community and Family Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Dolling JA, Boreham DR, Brown DL, Raaphorst GP, Mitchel RE. Cisplatin-modification of DNA repair and ionizing radiation lethality in yeast, Saccharomyces cerevisiae. Mutat Res 1999; 433:127-36. [PMID: 10102039 DOI: 10.1016/s0921-8777(98)00069-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cis-diamminedichloroplatinum II (cisplatin) is a DNA inter- and intrastrand crosslinking agent which can sensitize prokaryotic and eukaryotic cells to killing by ionizing radiation. The mechanism of radiosensitization is unknown but may involve cisplatin inhibition of repair of DNA damage caused by radiation. Repair proficient wild type and repair deficient (rad52, recombinational repair or rad3, excision repair) strains of the yeast Saccharomyces cerevisiae were used to determine whether defects in DNA repair mechanisms would modify the radiosensitizing effect of cisplatin. We report that cisplatin exposure could sensitize yeast cells with a competent recombinational repair mechanism (wild type or rad3), but could not sensitize cells defective in recombinational repair (rad52), indicating that the radiosensitizing effect of cisplatin was due to inhibition of DNA repair processes involving error free RAD52-dependent recombinational repair. The presence or absence of oxygen during irradiation did not alter this radiosensitization. Consistent with this result, cisplatin did not sensitize cells to mutation that results from lesion processing by an error prone DNA repair system. However, under certain circumstances, cisplatin exposure did not cause radiosensitization to killing by radiation in repair competent wild type cells. Within 2 h after a sublethal cisplatin treatment, wild type yeast cells became both thermally tolerant and radiation resistant. Cisplatin pretreatment also suppressed mutations caused by exposure to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), a response previously shown in wild type yeast cells following radiation pretreatment. Like radiation, the cisplatin-induced stress response did not confer radiation resistance or suppress MNNG mutations in a recombinational repair deficient mutant (rad52), although thermal tolerance was still induced. These results support the idea that cisplatin adducts in DNA interfere with RAD52-dependent recombinational repair and thereby sensitize cells to killing by radiation. However, the lesions can subsequently induce a general stress response, part of which is induction of RAD52-dependent error free recombinational repair. This stress response confers radiation resistance, thermal tolerance, and mutation resistance in yeast.
Collapse
Affiliation(s)
- J A Dolling
- Department of Biology, University of Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
24
|
Bibby MC, Thompson MJ, Rafferty JA, Margison GP, McElhinney RS. Influence of O6-benzylguanine on the anti-tumour activity and normal tissue toxicity of 1,3-bis(2-chloroethyl)-1-nitrosourea and molecular combinations of 5-fluorouracil and 2-chloroethyl-1-nitrosourea in mice. Br J Cancer 1999; 79:1332-9. [PMID: 10188873 PMCID: PMC2362701 DOI: 10.1038/sj.bjc.6690215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Previous studies have demonstrated that novel molecular combinations of 5-fluorouracil (5FU) and 2-chloroethyl-1-nitrosourea (CNU) have good preclinical activity and may exert less myelotoxicity than the clinically used nitrosoureas such as 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). This study examined the effect of O6-alkylguanine-DNA-alkyltransferase (ATase) depletion by the pseudosubstrate O6-benzylguanine (BG) on the anti-tumour activity and normal tissue toxicity in mice of three such molecular combinations, in comparison with BCNU. When used as single agents at their maximum tolerated dose, all three novel compounds produced a significant growth retardation of BCNU-resistant murine colon and human breast xenografts. This in vivo anti-tumour effect was potentiated by BG, but was accompanied by severe myelotoxicity as judged by spleen colony forming assays. However, while tumour resistance to BCNU was overcome using BG, this was at the expense of enhanced bone marrow, gut and liver toxicity. Therefore, although this ATase-depletion approach resulted in improved anti-tumour activity for all three 5-FU:CNU molecular combinations, the potentiated toxicities in already dose-limiting tissues indicate that these types of agents offer no therapeutic advantage over BCNU when they are used together with BG.
Collapse
Affiliation(s)
- M C Bibby
- Clinical Oncology Unit, University of Bradford, UK
| | | | | | | | | |
Collapse
|
25
|
Jenkins G, Takahashi N, Parry J. A study of ENU-induced mutagenesis in the mouse using the restriction site mutation (RSM) assay. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1520-6866(1999)19:4<281::aid-tcm5>3.0.co;2-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Garc�a-R�o L, Leis JR, Moreira JA, Norberto F. Stability and nitrosation efficiency of substitutedN-methyl-N-nitrosobenzenesulfonamides. J PHYS ORG CHEM 1998. [DOI: 10.1002/(sici)1099-1395(1998100)11:10<756::aid-poc48>3.0.co;2-d] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
27
|
Vogel EW, Nivard MJ, Ballering LA, Bartsch H, Barbin A, Nair J, Comendador MA, Sierra LM, Aguirrezabalaga I, Tosal L, Ehrenberg L, Fuchs RP, Janel-Bintz R, Maenhaut-Michel G, Montesano R, Hall J, Kang H, Miele M, Thomale J, Bender K, Engelbergs J, Rajewsky MF. DNA damage and repair in mutagenesis and carcinogenesis: implications of structure-activity relationships for cross-species extrapolation. Mutat Res 1996; 353:177-218. [PMID: 8692191 DOI: 10.1016/0027-5107(96)00032-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Previous studies on structure-activity relationships (SARs) between types of DNA modifications and tumour incidence revealed linear positive relationships between the log TD50 estimates and s-values for a series of mostly monofunctional alkylating agents. The overall objective of this STEP project was to further elucidate the mechanistic principles underlying these correlations, because detailed knowledge on mechanisms underlying the formation of genotoxic damage is an absolute necessity for establishing guidance values for exposures to genotoxic agents. The analysis included: (1) the re-calculation and further extension of TD50 values in mmol/kg body weight for chemicals carcinogenic in rodents. This part further included the checking up data for Swain-Scott s-values and the use of the covalent binding index (CBI); (2) the elaboration of genetic toxicity including an analysis of induced mutation spectra in specific genes at the DNA level, i.e., the vermilion gene of Drosophila, a plasmid system (pX2 assay) and the HPRT gene in cultured mammalian cells (CHO-9); and (3) the measurement of specific DNA alkylation adducts in animal models (mouse, rat, hamster) and mammalian cells in culture. The analysis of mechanisms controlling the expression of mammalian DNA repair genes (alkyltransferases, glycosylases) as a function of the cell type, differentiation stage, and cellular microenvironment in mammalian cells. The 3 classes of genotoxic carcinogens selected for the project were: (1) chemicals forming monoalkyl adducts upon interaction with DNA; (2) genotoxins capable of forming DNA etheno-adducts; and (3) N-substituted aryl compounds forming covalent adducts at the C8 position of guanine in DNA. In general, clear SARs and AARs (activity-activity relationships) between physiochemical parameters (s-values, O6/N7-alkylguanine ratios, CBI), carcinogenic potency in rodents and several descriptors of genotoxic activity in germ cells (mouse, Drosophila) became apparent when the following descriptors were used: TD50 estimates (lifetime doses expressed in mg/kg b.wt. or mmol/kg b.wt.) from cancer bioassays in rodents; the degree of germ-cell specificity, i.e., the ability of a genotoxic agent to induce mutations in practically all cell stages of the male germ-cell cycle of Drosophila (this project) and the mouse (literature search), as opposed to a more specific response in postmeiotic stages of both species; the Mexr-/Mexr+ hypermutability ratio, determined in a repair assay utilizing Drosophila germ cells; mutation spectra induced at single loci (the 7 loci used in the specific-locus test of the mouse (published data), and the vermilion gene of Drosophila); and doubling doses (DD) in mg/kg (mmol/kg) for specific locus test results on mice. By and large, the TD50 values, the inverse of which can be considered as measures of carcinogenic potency, were shown to be predictable from knowledge of the in vivo doses associated with the absorbed amounts of the investigated alkylators and with the second-order constant, kc, reaction at a critical nucleophilic strength, nc. For alkylating agents kc can be expressed as the second-order rate constant for hydrolysis, kH2O, and the substrate constant s:kH2OTD50 is a function of a certain accumulated degree of alkylation, here given as the (average) daily increment, ac, for 2 years exposure of the rodents. The TD*50 in mmol/kg x day) could then be written: [formula: see text] This expression would be valid for monofunctional alkylators provided the reactive species are uncharged. This is the case for most SN2 reagents. Although it appears possible to predict carcinogenic potency from measured in vivo doses and from detailed knowledge of reaction-kinetic parameter values, it is at present not possible to quantify the uncertainty of such predictions. One main reason for this is the complication due to uneven distribution in the body, with effects on the dose in target tissues. The estimation can be impro
Collapse
Affiliation(s)
- E W Vogel
- Department of Radiation Genetics and Chemical Mutagenesis, Medical Genetics Centre South-West Netherlands (MGC), University of Leiden (RUL), Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nivard MJ, Pastink A, Vogel EW. Mutational spectra induced under distinct excision repair conditions by the 3 methylating agents N-methyl-N-nitrosourea, N-methyl-N'-nitro-N-nitrosoguanidine and N-nitrosodimethylamine in postmeiotic male germ cells of Drosophila. Mutat Res 1996; 352:97-115. [PMID: 8676923 DOI: 10.1016/0027-5107(96)00011-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This paper describes the analysis of mutations induced at the vermilion locus in postmeiotic male germ cell stages of Drosophila exposed to 3 different N-methyl-N-nitroso compounds: N-methyl-N-nitrosourea (MNU); N-methyl-N'-nitro-N-nitrosoguanidine (MNNG); and N-nitrosodimethylamine (DMN). With MNU and DMN, the impact of DNA nucleotide excision repair (NER) on the spectra of mutations was studied. Mutants were isolated from F1 (mutations fixed before the first mitotic replication after fertilization) and F2 (mutations fixed following one or more mitotic replications; mosaics in F1) generations. The vermilion system enables the analysis of both intra- and inter-locus DNA changes for which several techniques have been adapted: (1) amplification of the vermilion gene by PCR, cloning of the fragment and sequence analysis of ssDNA; (2) Southern blot hybridization; and (3) cytological analysis of polytene chromosomes. In total, 49 MNU (26 from the exr+ genotype and 23 from the exr- genotype), 47 DMN (28 from the exr+ genotype and 19 from the exr- genotype) and 16 MNNG-induced mutations were characterized. The F1 spectra of all 3 agents contained base-pair changes and deletions (intra- and multi-locus) in a ratio of roughly 1 to 1, indicating a significant contribution of nitrogen DNA adducts to the spectra. In all F2 spectra the levels of base-pair changes were significantly higher compared to those in the F1 spectra, a finding also made for methyl methanesulfonate-induced mutations in earlier studies. There is an increase of mutations of, especially, the transversion types of mutations under exr- conditions in comparison to the exr+ situation. The induced transversions, clearly present in all spectra (exr+ and exr-), are presumably caused by N-methyl DNA adducts, which upon release from the DNA backbone lead to apurinic sites in a time-related process. Regarding the occurrence of transitions, it turned out for all 3 mutagens that the AT-->GC type strongly dominated the GC-->AT transitions. This suggest that O6-methylguanine is efficiently repaired, in contrast to O4-methylthymine. Based on the data obtained in the vermilion system with ENU, we propose, in addition, that the Drosophila alkyltransferase system repairs O6-methylguanine more efficiently than O6-ethylguanine.
Collapse
Affiliation(s)
- M J Nivard
- MGC-Department of Radiation Genetics and Chemical Mutagenesis, Leiden University, Sylvius Laboratories, Netherlands
| | | | | |
Collapse
|
29
|
Affiliation(s)
- M Hall
- Cancer Research Campaign Mammalian Cell DNA Repair Group, Department of Zoology, Cambridge, U.K
| | | |
Collapse
|
30
|
Wedge SR, Newlands ES. O6-benzylguanine enhances the sensitivity of a glioma xenograft with low O6-alkylguanine-DNA alkyltransferase activity to temozolomide and BCNU. Br J Cancer 1996; 73:1049-52. [PMID: 8624262 PMCID: PMC2074397 DOI: 10.1038/bjc.1996.203] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The effect of the O6-alkylguanine-DNA alkyltransferase (AGT) inhibitor, O6-benzylguanine (O6-BG), on the anti-tumour activity of 8-carbamoyl-3-methylimidazo [5,1-d]-1,2,3,5-tetrazine-4(3H)-one (temozolomide) or 1,3-bis(2-chloroethyl)-nitrosourea (BCNU) was evaluated in athymic mice bearing subcutaneous (s.c.) human glioma (U87MG) xenografts. The activity of AGT in U87MG xenografts was 4.3 +/- 1.5 fmol mg-1 protein (mean +/- s.d). These xenografts were inherently sensitive to treatment with alkylating compounds alone, with non-toxic doses of temozolomide (35 mg kg-1) or BCNU (10 mg kg-1) producing tumour growth delays of 23.3 and 11.8 days respectively. O6-BG (40 mg kg-1) did not inhibit tumour growth when administered alone, but was found to enhance significantly the anti-tumour activity of temozolomide or BCNU when administered 1 h before therapy (P < 0.002, Mann-Whitney test). AGT activity measured 24 h after the administration of 40 mg kg-1 O6-BG, was only 0.9 +/- 0.2 fmol mg-1 protein. These results are in contrast to previous studies in vitro with tumour cell lines of low AGT activity (< 15 fmol mg-1 protein), in which the cytotoxicity of temozolomide or BCNU was unaffected by AGT depletion.
Collapse
Affiliation(s)
- S R Wedge
- Department of Medical Oncology, Charing Cross Hospital, London, UK
| | | |
Collapse
|
31
|
Wei YF, Carter KC, Wang RP, Shell BK. Molecular cloning and functional analysis of a human cDNA encoding an Escherichia coli AlkB homolog, a protein involved in DNA alkylation damage repair. Nucleic Acids Res 1996; 24:931-37. [PMID: 8600462 PMCID: PMC145711 DOI: 10.1093/nar/24.5.931] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The Escherichia coli AlkB protein is involved in protecting cells against mutation and cell death induced specifically by SN2-type alkylating agents such as methyl methanesulfonate (MMS). A human cDNA encoding a polypeptide homologous to E.coli AlkB was discovered by searching a database of expressed sequence tags (ESTs) derived from high throughput cDNA sequencing. The full-length human AlkB homolog (hABH) cDNA clone contains a 924 bp open reading frame encoding a 34 kDa protein which is 52% similar and 23% identical to E.coli AlkB. The hABH gene, which maps to chromosome 14q24, was ubiquitously expressed in 16 human tissues examined. When hABH was expressed in E.coli alkB mutant cells partial rescue of the cells from MMS-induced cell death occurred. Under the conditions used expression of hABH in skin fibroblasts was not regulated by treatment with MMS. Our findings show that the AlkB protein is structurally and functionally conserved from bacteria to human, but its regulation may have diverged during evolution.
Collapse
Affiliation(s)
- Y F Wei
- Department of Molecular Biology, Human Genome Sciences Inc., Rockville, MD 20850-3338, USA
| | | | | | | |
Collapse
|
32
|
Mineura K, Watanabe K, Yanagisawa T, Kowada M. Quantification of O6-methylguanine-DNA methyltransferase mRNA in human brain tumors. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1289:105-9. [PMID: 8605218 DOI: 10.1016/0304-4165(95)00123-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
O6-Methylguanine-DNA methyltransferase (MGMT) is strongly involved in drug resistance mechanism of tumor cells to chloroethylnitrosoureas (CENUs), because it removes and repairs CENU-induced O6-alkylguanine-DNA by accepting the alkyl group at a cysteine moiety. MGMT activity and MGMT mRNA expression are good indicators for detection of sensitive cells or resistant cells to CENUs. In the present study, we applied a non-radioactive reverse transcription-polymerase chain reaction (RT-PCR) method on quantitative measurement of MGMT mRNA expression. Estimated levels of MGMT mRNA expression determined by this RT-PCR method were consistent with the actual doses of MGMT mRNA. This relationship was noted at a wide range from 10 fg to 10 pg. The relative expression levels of MGMT mRNA estimated from kinetic analysis correlated well with MGMT activity determined using 3H-methyl-nitrosourea-treated DNA substrate in brain tumor cells (P<0.001 with a correlation coefficient of 0.997). The RT-PCR method facilitated quantitative measurements in even a small amount of biopsy specimens obtained by stereotactic brain surgery.
Collapse
Affiliation(s)
- K Mineura
- Neurosurgical Service, Akita University Hospital, Japan
| | | | | | | |
Collapse
|
33
|
Wedge SR, Porteus JK, May BL, Newlands ES. Potentiation of temozolomide and BCNU cytotoxicity by O(6)-benzylguanine: a comparative study in vitro. Br J Cancer 1996; 73:482-90. [PMID: 8595163 PMCID: PMC2074446 DOI: 10.1038/bjc.1996.85] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Depletion of the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) with O(6)-benzylguanine (O(6)-BG) has been widely shown to enhance 1,3-bis(2-chloroethyl)-nitrosourea (BCNU) activity. This study aimed to determine whether temozolomide, a methylating imidazotetrazinone, would similarly benefit from combination with O(6)-BG. Seven human cell lines were examined with AGT activities ranging from <6 fmol mg-1 protein to >700 fmol mg-1 protein. Comparisons with BCNU were made on both single and multiple dosing schedules, since temozolomide cytotoxicity is highly schedule dependent. In single-dose potentiation studies, cells were preincubated with 100 microM O(6)-BG for 1 h, a treatment found to deplete AGT activity by >90% for 24 h. No potentiation of either temozolomide or BCNU cytotoxicity was observed in two glioblastoma cell lines with <6 fmol mg-1 protein AGT. In all other cell lines studied potentiation of BCNU toxicity by O(6)-BG was between 1.6- and 2.3-fold and exceeded that of temozolomide (1.1- to 1.7-fold). The magnitude of this potentiation was unrelated to AGT activity and the relative potentiation of temozolomide and BCNU cytotoxicity was found to be highly variable between cell lines. In multiple dosing studies two colorectal cell lines (Mawi and LS174T) were treated with temozolomide or BCNU at 24 h intervals for up to 5 days, with or without either 100 microM O(6)-BG for 1 h or 1 microM O(6)-BG for 24 h, commencing 1 h before alkylating treatment. Extended treatment with 1 microM O(6)-BG produced greater potentiation than intermittent treatment with 100 microM O(6)-BG. Potentiation of temozolomide cytotoxicity increased linearly in Mawi with each subsequent dosing: from 1.4-fold (day 1) to 4.2-fold (day 5) with continuous 1 microM O(6)-BG. In contrast, no potentiation was observed in LS174T, a cell line that would appear to be 'tolerant' of methylation. Potentiation of BNCU cytotoxicity increased in both cell lines with repeat dosing, although the rate of increase was less than that observed with temozolomide and continuous 1 microM O(6)-BG in Mawi. These results suggest that repeat dosing of an AGT inhibitor and temozolomide may have a clinical role in the treatment of tumours that exhibit AGT-mediated resistance.
Collapse
Affiliation(s)
- S R Wedge
- Department of Medical Oncology, Charing Cross Hospital, London, UK
| | | | | | | |
Collapse
|
34
|
Mineura K, Fukuchi M, Kowada M, Terashima I, Kohda K. Differential inactivation of O6-methylguanine-DNA methyltransferase activity by O6-arylmethylguanines. Int J Cancer 1995; 63:148-51. [PMID: 7558444 DOI: 10.1002/ijc.2910630126] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Activity of O6-methylguanine-DNA methyltransferase (MGMT) is well related with drug resistance of tumor cells to chloroethylnitrosoureas (CENUs), because MGMT removes CENU-induced O6-alkylguanines in DNA by accepting the alkyl group at a cysteine moiety. Inactivation of MGMT is a feasible way to overcome MGMT-related resistance of tumor cells to CENUs. O6-Benzylguanine is known to be a strong depleter of MGMT. We previously reported the potentiation effect of O6-arylmethylguanine derivatives on cytotoxicity of 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride (ACNU), a CENU, for HeLa S3 cells. In this study, we tested the activity of these O6-arylmethylguanine derivatives as depleters of MGMT using HeLa S3 cell-free extract. The O6-arylmethylguanine derivatives tested were O6-benzylguanine (1), O6-(4-, 3-, and 2-fluorobenzyl)guanines (2-4), O6-(4, 3-, and 2-trifluoromethylbenzyl)guanines (5-7), O6-(4-, 3-, and 2-pyridylmethyl)guanines (8-10), and O6-(2- and 1-naphtylmethyl)guanines (11,12). Among these, compounds 1-3, 5, 8, 9 and 11 showed a strong MGMT depletion activity, whereas compounds 4, 6, 7, 10 and 12 were inactive. These inactive compounds, except for 6, have a substituent at the alpha position of the benzyl group (4, 7, 12) or are an alpha nitrogen analogue of 1 (10). There was a good relation (r = -0.856, p < 0.001) between the MGMT depletion activity of O6-arylmethylguanine derivatives and their potentiation activity of ACNU cytotoxicity. These results suggest that the alpha position of the benzyl group plays an important role in the interaction of O6-arylmethylguanine derivatives with MGMT to result in the inactivation of MGMT. Potent MGMT inactivators (1-3, 5, 8, 9, 11) sensitize tumor cells to CENU chemotherapy.
Collapse
Affiliation(s)
- K Mineura
- Neurosurgical Service, Akita University Hospital, Japan
| | | | | | | | | |
Collapse
|
35
|
Abstract
Alkylation-induced germ cell mutagenesis in the mouse versus Drosophila is compared based on data from forward mutation assays (specific-locus tests in the mouse and in Drosophila and multiple-locus assays in the latter species) but not including assays for structural chromosome aberrations. To facilitate comparisons between mouse and Drosophila, forward mutation test results have been grouped into three categories. Representatives of the first category are MMS (methyl methanesulfonate) and EO (ethylene oxide), alkylating agents with a high s value which predominantly react with ring nitrogens in DNA. ENU (N-ethyl-N-nitrosourea), MNU (N-methyl-N-nitrosourea), PRC (procarbazine), DEN (N-nitrosodiethylamine), and DMN (N-nitrosodimethylamine) belong to the second category. These agents have in common a considerable ability for modification at oxygens in DNA. Cross-linking agents (melphalan, chlorambucil, hexamethylphosphoramide) form the third category. The most unexpected, but encouraging outcome of this study is the identification of common features for three vastly different experimental indicators of genotoxicity: hereditary damage in Drosophila males, genetic damage in male mice, and tumors (TD50 estimates) in rodents. Based on the above three category classification scheme the following tentative conclusions are drawn. Monofunctional agents belonging to category 1, typified by MMS and EO, display genotoxic effects in male germ cell stages that have passed meiotic division. This phenomenon seems to be the consequence of a repair deficiency during spermiogenesis for a period of 3-4 days in Drosophila and 14 days in the mouse. We suggest that the reason for the high resistance of premeiotic stages, and the generally high TD50 estimates observed for this class in rodents, is the efficient error-free repair of N-alkylation damage. If we accept this hypothesis, then the increased carcinogenic potential in rodents, seen when comparing category 2 (ENU-type mutagens) to category 1 (MMS-type mutagens), along with the ability of category 2 genotoxins to induce genetic damage in premeiotic stages, must presumably be due to their enhanced ability for alkylations at oxygens in DNA; it is this property that actually distinguishes the two groups from each other. In contrast to category 1, examination of class 2 genotoxins (ENU and DEN) in premeiotic cells of Drosophila gave no indication for a significant role of germinal selection, and also removal by DNA repair was less dramatic compared to MMS. Thus category 2 mutagens are expected to display activity in a wide range of both post- and premeiotic germ cell stages. A number of these agents have been demonstrated to be among the most potent carcinogens in rodents.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- E W Vogel
- Medical Genetics Centre South-West Netherlands-MGC, Department of Radiation Genetics and Chemical Mutagenesis, Leiden University
| | | |
Collapse
|
36
|
Bobola MS, Blank A, Berger MS, Silber JR. Contribution of O6-methylguanine-DNA methyltransferase to monofunctional alkylating-agent resistance in human brain tumor-derived cell lines. Mol Carcinog 1995; 13:70-80. [PMID: 7605582 DOI: 10.1002/mc.2940130203] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) has been implicated in resistance of human brain tumors to alkylating agents. We observed that 14 human medulloblastoma- and glioma-derived cell lines differ in sensitivity to the methylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), as shown by their 28-fold range in 10% survival dose (LD10). By using the substrate analogue inhibitor O6-benzylguanine (O6-BG), we showed that the contribution of MGMT to resistance varies widely, as evidenced by 3- to 30-fold reductions in LD10 among the lines, and varies up to 20-fold among subpopulations of individual lines. Importantly, variability in resistance, manifested as a 20-fold range in LD10, persists after measurable MGMT is eliminated, disclosing differential contributions of other resistance mechanisms to survival. Cells exposed to MNNG while suspended in growth medium are more resistant than cells alkylated as subconfluent monolayers, and MGMT accounts for a smaller proportion of their resistance. Notably, the MGMT content of the lines is not statistically correlated with MNNG resistance or with potentiation of killing by O6-BG, even though MGMT is a biochemically demonstrated determinant of resistance. In contrast, the same lines vary less in resistance to the ethylating agent N-ethylnitrosourea (ENU), and MGMT makes only a small contribution to resistance. Our results strongly indicate that resistance to both MNNG and ENU is multifactorial.
Collapse
Affiliation(s)
- M S Bobola
- Department of Neurological Surgery, University of Washington, Seattle 98195, USA
| | | | | | | |
Collapse
|
37
|
Xiao W, Fontanie T. Expression of the human MGMT O6-methylguanine DNA methyltransferase gene in a yeast alkylation-sensitive mutant: its effects on both exogenous and endogenous DNA alkylation damage. Mutat Res 1995; 336:133-42. [PMID: 7885384 DOI: 10.1016/0921-8777(94)00048-b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Common Mer- cell lines deficient in O6-methylguanine DNA methyltransferase (MTase) activity probably result from the down-regulation of, rather than mutations in, the MGMT gene. However, the down-regulation of other unrelated genes was also observed in some of these cell lines, making it difficult to determine the precise functions of the MGMT MTase gene. To study the biological function of human MGMT MTase, we seek to utilize a newly created yeast mgt1 mutant deficient in the DNA repair MTase activity. The human MGMT cDNA was cloned into yeast expression vectors so that the MGMT gene is under the control of either an inducible GAL1 promoter or a constitutive ADH1 promoter. Upon galactose induction, the PGAL1-MGMT transformant had about 40-fold MTase activity compared to the wild-type strain. MGMT overexpression protected the yeast mgt1 mutant against alkylation-induced killing and mutation. Limited expression of the MGMT gene in the mgt1 mutant still provides significant alkylation resistance, albeit at a reduced level. The yeast mgt1 mutants increase spontaneous mutation rate, whereas constitutive expression of the MGMT gene lowered the spontaneous mutation rate in the mgt1 mutant to the wild-type level. We suggest that MGMT MTase may play the same role in human cells as the MGT1 MTase in yeast cells. Thus our results demonstrate that the human MGMT gene functionally complements the yeast MTase-deficient mutant in the protection against exogenous and endogenous DNA alkylation damage, which provides a useful tool for the study of in vivo mammalian MTase functions.
Collapse
Affiliation(s)
- W Xiao
- Department of Microbiology, University of Saskatchewan, Saskatoon, Canada
| | | |
Collapse
|
38
|
Sledziewska-Gójska E. Inactivation of O6-methylguanine-DNA methyltransferase in vivo by SN2 alkylating agents. Mutat Res 1995; 336:61-7. [PMID: 7528896 DOI: 10.1016/0921-8777(94)00038-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The cellular level of O6-methylguanine-DNA methyltransferase (MGMT) is important in mutagenic, carcinogenic and therapeutic effects of alkylating agents. I have investigated how SN2 alkylating agents affect the activity of MGMT in vivo. As a model, adapted cultures of E. coli K12 strain AB2497 containing 2400 +/- 430 molecules of MGMT per cell were used. MGMT activity was assayed in the cell extracts of adapted cultures challenged with various doses of MMS, DMS and for comparison the SN1 alkylating agents, MNNG and MNU. In control non-adapted cultures, with low constitutive levels of MGMT, the mutagenic potential of various doses of different alkylating agents was estimated to correlate with the O6-methylguanine content produced in DNA by various treatments. Inactivation of MGMT by MNNG or MNU occurs only in doses able to produce a highly mutagenic level of O6-methylguanine in DNA, which is consistent with the consumption of MGMT activity in the DNA repair process. In contrast, non-mutagenic doses of MMS or DMS are sufficient to inactivate MGMT in adapted E. coli cells. It may be concluded that SN2 alkylating agents can block the main pathway of O6-methylguanine-DNA repair in vivo.
Collapse
|
39
|
Wilson DM, Tentler JJ, Carney JP, Wilson TM, Kelley MR. Acute ethanol exposure suppresses the repair of O6-methylguanine DNA lesions in castrated adult male rats. Alcohol Clin Exp Res 1994; 18:1267-71. [PMID: 7847618 DOI: 10.1111/j.1530-0277.1994.tb00117.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alcohol has clearly been associated with an increase of cancers in numerous tissue, including the respiratory tract, colon, rectum, liver, but especially the esophagus, larynx, pharynx, and mouth. Alcohol alone has not been shown to be a mutagen until it is converted to acetaldehyde and, therefore, alcohol presumably acts as a cocarcinogen. Previous data has shown that alcohol concentrations of 2% or greater inhibits DNA repair, and in light of the widespread consumption of alcoholic beverages with alcohol contents ranging from 4 to 5% (beer and wine coolers) to 50% (whiskey), interest in determining the mechanism(s) responsible for alcohol-induced carcinogenesis has heightened. Although previous studies, in intact rats, have investigated the effects of chronic alcohol exposure on some aspects of DNA repair, we have begun to address the effects of acute or "binge" alcohol exposure on mammalian DNA repair. Toward this end, we report the inhibition of O6-methylguanine-DNA methyltransferase (MGMT) by a single intraperitoneal injection of 30% ethanol in adult male castrated rats. This inhibition lasted for at least 24 hr. We also observed a dose-response effect of ethanol on MGMT activity, again only in the castrated rats. The finding of ethanol's effect on MGMT activity in castrated and not intact rats implies a hormonal component of MGMT DNA repair response, which has only been alluded to in past research.
Collapse
Affiliation(s)
- D M Wilson
- Department of Pediatrics, Riley Hospital, Indianapolis, IN 46202
| | | | | | | | | |
Collapse
|
40
|
Abstract
Resistance to chemotherapy in brain tumors is complex and may involve multiple mechanisms. For commonly used drugs, such as nitrosoureas and platinum compounds, major mechanisms may involve increaded DNA repair or removal of the drug-DNA adducts. For water soluble nitrosoureas and also for platinum compounds, other mechanisms, such as alteration in drug transport, may be important. Another major mechanism may involve glutathione and glutathione-S-transferase pathways. For vinca alkaloids and epipodophyllotoxins p-glycoprotein mediated MDR appears to be the major feature in drug resistance. In addition, alteration of tubulin and topoisomerase II have been described in resistance to vinca alkaloids and epipodophyllotoxins respectively. Recently, increased multidrug resistance associated protein gene expression has been found in glioma cells and brain tumor samples; its clinical significance requires further investigation.
Collapse
Affiliation(s)
- L G Feun
- University of Miami Hospital and Clinics, Florida
| | | | | |
Collapse
|
41
|
Burkhart JG, Burkhart BA, Sampson KS, Malling HV. ENU-induced mutagenesis at a single A: T base pair in transgenic mice containing phi X174. Mutat Res 1993; 292:69-81. [PMID: 7688099 DOI: 10.1016/0165-1161(93)90009-o] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Transgenic mice containing the bacteriophage phi X174 am3 as a chromosomally integrated and recoverable marker for in vivo mutation have been produced to measure spontaneous and induced substitutions at an A:T base pair among single copies. phi X174 was chosen for its small size (5 kb), unique sequence, and the opportunity to take advantage of previously reported in vitro data on mutation and repair; the am3 site provides sequence specificity in a reversion assay for mutation of an A:T base pair. Inbred C57Bl/6 mice have been made homozygous for approximately 100 copies of the the phage sequence without any apparent detrimental effects on the homozygous individuals. Recoveries of phage from mouse tissues are in the range of 1-5 x 10(7) PFU per micrograms mouse DNA; both recovery and mutation are independent of endogenous CpG methylation. Background mutation frequencies are 2-4 x 10(-7) among phage recovered from liver, brain, spleen, and kidney. Adult mice were treated with 200 mg/kg N-ethyl-N-nitrosourea, and phage were recovered at 2 and 14 days after treatment. At 2 days after treatment we observed a slight increase only among phage isolated from the brain of one mouse out of four. At 14 days after ENU treatment, there were significant increases in mutation frequencies among phage recovered from the liver (6 x) and spleen (10 x). These results demonstrate (1) response of a single A:T base pair to alkylation-induced mutation in a nonexpressed gene, (2) the role of cell proliferation in somatic mutagenesis, and (3) provide a model for a transgenic approach for study of site-specific mutagenesis in vivo in higher eukaryotes.
Collapse
Affiliation(s)
- J G Burkhart
- Laboratory of Genetics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| | | | | | | |
Collapse
|
42
|
Sledziewska-Gójska E. The level of GC-->AT transitions induced by MMS is not affected by the adaptive response in Escherichia coli K12. Mutat Res 1993; 294:1-8. [PMID: 7683754 DOI: 10.1016/0921-8777(93)90052-i] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
I have examined the effect of the adaptive response on the frequency of MMS-induced umuC-dependent AT-->TA transversions and umuC-independent, GC-->AT transitions. It was found that the induction of the adaptive response causes a moderate decrease (50-60%) in the frequency of AT-->TA transversions and surprisingly has no effect on the level of GC-->AT transitions. In contrast, a dramatic decrease in MNNG-induced mutations has been observed in adapted cultures. However, this effect was completely abolished by MMS pretreatment of adapted cells before MNNG challenge. A mechanism for the MMS interference with the repair of MNNG-induced mutations in adapted cells is proposed.
Collapse
|
43
|
Mizoguchi M, Naito H, Kurata Y, Shibata MA, Tsuda H, Wild CP, Montesano R, Fukushima S. Influence of aging on multi-organ carcinogenesis in rats induced by N-methyl-N-nitrosourea. Jpn J Cancer Res 1993; 84:139-46. [PMID: 8463131 PMCID: PMC5919123 DOI: 10.1111/j.1349-7006.1993.tb02847.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The effects of aging on the multi-organ carcinogenesis induced in rats by N-methyl-N-nitrosourea (MNU), a direct carcinogen which does not need metabolic activation to exert carcinogenicity, were examined in male F344 rats. In the first experiment, rats at 6, 52, and 98 weeks of age were treated with MNU (20 mg/kg body weight, i.p.) twice weekly for 6 weeks and then maintained without any further treatment for 24 weeks in the case of young and middle-aged rats and for 18 weeks in the case of the old rats. In young rats, malignant lymphomas, particularly thymic types, were observed at significantly high incidence. A striking result in the middle-aged rats was the significantly higher incidence of adenocarcinomas in the small intestine than in young or old animals. The induction of proliferative and neoplastic lesions of the large intestine also tended to be increased in middle-aged rats. In addition, epithelial hyperplasia of the tongue, but not the forestomach, occurred at the highest incidence in the middle-aged group. There were no differences in the induction of epithelial lesions in the urinary bladder among the groups. In a second experiment, investigation of DNA synthesis in the tongue, small and large intestines, urinary bladder and lymph nodes did reveal significant increases or tendency for increase in the MNU-treated groups, but without differences with age. In contrast, the thymus of young rats showed significantly increased incorporation of BrdU label after administration of MNU, whereas it was markedly reduced in middle-aged rats. In a third experiment, O6-methyldeoxyguanine (O6-medG) DNA adduct formation was immunohistochemically detected in various organs including the thymus, forestomach, and small intestine without any differences with age. Thus, the results demonstrated that while the target organs of MNU are modified by the age of the animals, levels of DNA synthesis and O6-medG DNA adduct formation in most cases can not explain the observed differences in carcinogenic susceptibility.
Collapse
Affiliation(s)
- M Mizoguchi
- First Department of Pathology, Nagoya City University Medical School
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Felker GM, Friedman HS, Dolan ME, Moschel RC, Schold C. Treatment of subcutaneous and intracranial brain tumor xenografts with O6-benzylguanine and 1,3-bis(2-chloroethyl)-1-nitrosourea. Cancer Chemother Pharmacol 1993; 32:471-6. [PMID: 8258196 DOI: 10.1007/bf00685892] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
O6-Alkylguanine-DNA alkyltransferase (AT) is a cellular protein that protects cells from the cytotoxic effects of nitrosoureas by repairing alkyl lesions at the O6 position of guanine. We have studied the ability of O6-benzylguanine to deplete AT activity in brain tumor xenografts and thereby increase the sensitivity of these tumors to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). In toxicity studies, pretreatment of athymic mice with O6-benzylguanine increased the toxicity of BCNU significantly. After i.p. injection of O6-benzylguanine into athymic mice carrying subcutaneous (s.c.) D341MED, a human medulloblastoma xenograft with a high AT activity, the AT activity of the tumors became undetectable within 1 h and remained depleted until 36 h. In s.c. xenografts to D341MED, treatment with O6-benzylguanine followed 1 h later by BCNU produced a significantly greater growth delay (14.8 days) than was seen with BCNU alone (2.3 days). A lower pretreatment dose of O6-benzylguanine produced a significantly smaller therapeutic effect. Delaying the administration of BCNU until 36 h after O6-benzylguanine resulted in a growth delay (1.2 days) that was not significantly different from that produced by the control or BCNU alone. In athymic mice with intracranial (i.c.) xenografts of D341MED, pretreatment with O6-benzylguanine followed 1 h later by BCNU produced a significantly increased survival as compared with that of the control, BCNU alone, O6-benzylguanine alone, and O6-benzylguanine followed 36 h later by BCNU. In experiments with s.c. xenografts of D245MG, a human glioma xenograft with undetectable AT activity, pretreatment with O6-benzylguanine 1 h prior to BCNU produced a significantly greater effect than was seen with BCNU treatment alone. The combination regimen, however, was not as effective as an equitoxic dose of BCNU alone. These studies suggest that O6-benzylguanine may be a useful adjuvant to nitrosourea therapy in human malignancies that exhibit a range of AT activities and that dose and timing are important variables in achieving therapeutic success. These data also indicate that therapeutic potentiation of BCNU by O6-benzylguanine can be achieved in i.c. tumors. As a result, this approach may be useful in the treatment of neoplasms of the central nervous system.
Collapse
Affiliation(s)
- G M Felker
- Duke University School of Medicine, Durham, NC 27710
| | | | | | | | | |
Collapse
|
45
|
Sarkar A, Dolan ME, Gonzalez GG, Marton LJ, Pegg AE, Deen DF. The effects of O6-benzylguanine and hypoxia on the cytotoxicity of 1,3-bis(2-chloroethyl)-1-nitrosourea in nitrosourea-resistant SF-763 cells. Cancer Chemother Pharmacol 1993; 32:477-81. [PMID: 8258197 DOI: 10.1007/bf00685893] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
O6-Alkylguanine-DNA alkyltransferase (AGT) activity is associated with resistance of brain tumor cell lines to the cytotoxic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). SF-763 cells exhibit high AGT activity and are resistant to BCNU. In this study, we compared the effects of the AGT inhibitor O6-benzylguanine (BG) on the cytotoxicity of BCNU in oxic and hypoxic SF-763 cells; we also measured AGT activity, ornithine decarboxylase (ODC) activity, and polyamine levels to determine if there was any correlation with cell survival as determined by colony-forming efficiency assay. Exponentially growing monolayer cells were pretreated with 10 microM BG for 2 h under oxic or hypoxic (95% nitrogen/5% CO2) conditions and then exposed to graded concentrations of BCNU for 1 h. BG significantly lowered AGT activity but had no cytotoxic effect in oxic or hypoxic cells; hypoxia alone was not cytotoxic. The cytotoxicity of BCNU was 4 times higher in BG-treated hypoxic cells than in oxic cells treated with BCNU alone; the BCNU doses required for a 1-log cell kill were 75 and 300 microM, respectively. ODC activity was lowered by hypoxia alone but was not significantly affected by BG in either hypoxic or oxic cells. Polyamine levels were not significantly affected by hypoxia or BG. These results indicate that pretreatment with BG dramatically lowers AGT activity and increases the cytotoxicity of BCNU in both oxic and hypoxic SF-763 cells. The mechanism of this enhanced cytotoxicity is apparently unrelated to ODC activity or polyamine levels.
Collapse
Affiliation(s)
- A Sarkar
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco 94143
| | | | | | | | | | | |
Collapse
|
46
|
Burkhart JG, Malling HV. Mutagenesis and transgenic systems: perspective from the mutagen, N-ethyl-N-nitrosourea. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 1993; 22:1-6. [PMID: 8339722 DOI: 10.1002/em.2850220103] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Affiliation(s)
- J G Burkhart
- National Institute Environmental Health Sciences, Research Triangle Park, NC 27709
| | | |
Collapse
|
47
|
Abstract
The etiologic factors responsible for IBD remain only speculative. It does appear that the inappropriate activation of the immune system, whether by immune complex deposition, infectious agents or vascular impairment, is important in the pathogenesis of these diseases. Interaction of certain cytokines known to be produced in human IBD with specific immune cells such as neutrophils and macrophages results in the induction of the enzyme NO. synthase with the concomitant release of large amounts of NO.. Nitric oxide is known to mediate many of the pathophysiological alterations associated with IBD including cell injury, intestinal hyperemia and intestinal smooth muscle dysfunction. In addition, NO. is known to decompose in solution to yield potent N-nitrosating agents which will N-nitrosate certain amines to yield potent carcinogenic nitrosamines. Because antioxidants (including 5-ASA) are potent inhibitors of nitrosamine formation, they may prove useful in attenuating the formation of potentially carcinogenic agents in vivo.
Collapse
Affiliation(s)
- M B Grisham
- Department of Physiology and Biophysics, Louisiana State University Medical Center, Shreveport 71130
| | | |
Collapse
|
48
|
Pardini C, Piras A, Voliani M, Rainaldi G, Mariani L, Taverna P, D'Incalci M, Citti L. Chinese hamster ovary cells deficient or proficient in O6-alkylguanine-DNA alkyltransferase activity are equally sensitive to X-rays. Mutat Res 1992; 283:125-9. [PMID: 1381489 DOI: 10.1016/0165-7992(92)90144-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In mammalian cells, under aerobic conditions, ionizing radiations and radiomimetic chemical agents can induce an enzymatic activity involved in DNA repair, O6-alkylguanine-DNA alkyltransferase (O6-AT). This catalytic protein is active against alkyl-radical-induced DNA damages. This induction was proposed to be linked to the formation of hydroxyl radicals. The possible involvement of O6-AT in the defense mechanism of the cell against aerobic radiation damage was investigated by comparing the X-ray sensitivity of two Chinese hamster ovary (CHO) cell lines, the first deficient (CHO mex-) and the second proficient by transfection of O6-AT (CHO mex+). The colony-forming ability after X-irradiation was appreciably reduced in CHO mex- in comparison to CHO mex+ cells. Nevertheless, pretreatment of proficient cells with O6-methylguanine, a specific inhibitor of O6-AT, reduced the DNA repair activity but did not modify the degree of sensitivity to X-rays of the CHO mex+ cells. Since the glutathione concentrations as well as the DNA damage amounts induced by X-irradiation were comparable in the variously treated cell lines, these results suggest that the observed induction of O6-AT by ionizing radiation in aerobic conditions could be a generalized rather than a specific response to damage by radicals.
Collapse
Affiliation(s)
- C Pardini
- Genetica e Biochemica Tossicologica dell'Istituto di Mutagenesi e Differenziamento-CNR, Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Osmak M, Horvat D. Chromosomal analysis of Chinese hamster V79 cells exposed to multiple gamma-ray fractions: induction of adaptive response to mitomycin C. Mutat Res 1992; 282:259-63. [PMID: 1379688 DOI: 10.1016/0165-7992(92)90132-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chinese hamster V79 cells were irradiated daily with 0.3 Gy of gamma-rays 5 times per week for 12 weeks (total 18 Gy). These cells were challenged with an additional dose of 15. Gy gamma-rays or treated with 5 micrograms/ml of mitomycin C (MMC) for 2 h. In spite of the high total accumulated dose of gamma-rays, the number of chromosomal aberrations and sister-chromatid exchanges (SCEs) did not significantly increase in the preirradiated cells, as compared to control cells. If preirradiated cells were challenged with an additional 1.5 Gy of gamma-rays, an insignificant decrease in the yield of chromatid aberrations was observed. In contrast, preirradiated cells became significantly more resistant to the induction of chromosomal damage when challenged with mitomycin C. Our results suggest that multiple fractions of gamma-rays can induce the adaptive response to mitomycin C in preirradiated cells.
Collapse
Affiliation(s)
- M Osmak
- Department of Experimental Biology and Medicine, Ruder Bosković Institute, Zagreb, Croatia
| | | |
Collapse
|
50
|
Rafferty JA, Elder RH, Watson AJ, Cawkwell L, Potter PM, Margison GP. Isolation and partial characterisation of a Chinese hamster O6-alkylguanine-DNA alkyltransferase cDNA. Nucleic Acids Res 1992; 20:1891-5. [PMID: 1579490 PMCID: PMC312303 DOI: 10.1093/nar/20.8.1891] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The cDNA encoding Chinese hamster O6-alkylguanine-DNA-alkyltransferase (ATase) has been isolated from a library prepared from RNA isolated from V79 lung fibroblasts which had an upregulated level of this repair activity following stepwise selection with a chloroethylating agent (1, 2). Expression of the cDNA in E. coli produced functionally active ATase at levels of 2.5% of total cellular protein as determined by in vitro assay. The recombinant hamster protein has a molecular weight of 28 kDa as estimated by SDS-PAGE and fluorography and this was identical to that in the upregulated cells. The characteristic PCHRV pentapeptide of the alkyl acceptor site has been identified and there is a 68 amino acid residue region which is 90% conserved across all the mammalian proteins so far analysed: in contrast, the N- and C-terminal domains diverge by as much as 50% between species. Polyclonal antibodies to the human and rat ATases hybridised to the hamster protein on western analysis suggesting at least one common epitope shared across species. However, in antibody inhibition experiments neither of the antisera cross reacted with the hamster ATase in a way which interfered with functional activity whereas the anti-human antibodies inhibited the human ATase and the anti-rat antibodies inhibited the rat and mouse ATases. There may therefore be significant tertiary structural differences between the hamster protein and the other mammalian ATases.
Collapse
Affiliation(s)
- J A Rafferty
- CRC Department of Chemical Carcinogenesis, Paterson Institute for Cancer Research, Christie Hospital, Manchester, UK
| | | | | | | | | | | |
Collapse
|