Elsevier

DNA Repair

Volume 6, Issue 8, 1 August 2007, Pages 1079-1099
DNA Repair

MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents

https://doi.org/10.1016/j.dnarep.2007.03.008Get rights and content

Abstract

O6-methylguanine-DNA methyltransferase (MGMT) plays a crucial role in the defense against alkylating agents that generate, among other lesions, O6-alkylguanine in DNA (collectively termed O6-alkylating agents [O6AA]). The defense is highly important, since O6AA are common environmental carcinogens, are formed endogenously during normal cellular metabolism and possibly inflammation, and are being used in cancer therapy. O6AA induced DNA damage is subject to repair, which is executed by MGMT, AlkB homologous proteins (ABH) and base excision repair (BER). Although this review focuses on MGMT, the mechanism of repair by ABH and BER will also be discussed. Experimental systems, in which MGMT has been modulated, revealed that O6-methylguanine (O6MeG) and O6-chloroethylguanine are major mutagenic, carcinogenic, recombinogenic, clastogenic and killing lesions. O6MeG-induced clastogenicity and cell death require MutSα-dependent mismatch repair (MMR), whereas O6-chloroethylguanine-induced killing occurs independently of MMR. Extensive DNA replication is required for O6MeG to provoke cytotoxicity. In MGMT depleted cells, O6MeG induces apoptosis almost exclusively, barely any necrosis, which is presumably due to the remarkable ability of secondarily formed DNA double-strand breaks (DSBs) to trigger apoptosis via ATM/ATR, Chk1, Chk2, p53 and p73. Depending on the cellular background, O6MeG activates both the death receptor and the mitochondrial apoptotic pathway. The inter-individual expression of MGMT in human lymphocytes is highly variable. Given the key role of MGMT in cellular defense, determination of MGMT activity could be useful for assessing a patient's drug sensitivity. MGMT is expressed at highly variable amounts in human tumors. In gliomas, a correlation was found between MGMT activity, MGMT promoter methylation and response to O6AA. Although the human MGMT gene is inducible by glucocorticoids and genotoxins such as radiation and alkylating agents, the role of this induction in the protection against carcinogens and the development of chemotherapeutic alkylating drug resistance are still unclear. Modulation of MGMT expression in tumors and normal tissue is currently being investigated as a possible strategy for improving cancer therapy.

Introduction

Mutagens in the environment [1], in tobacco smoke [2] and food [3], as well as endogenous metabolic products [4] generate reactive electrophilic species that alkylate DNA. Despite their carcinogenic potential, monofunctional alkylating agents are used, due to their cytotoxic properties, in chemotherapy of various cancers such as Hodgkin's disease, non-Hodgkin's lymphoma, malignant melanoma, neuroblastoma, soft tissue sarcomas, pancreatic (islet cell) cancer, carcinoid tumors, astrocytoma, glioblastoma and brain metastasis from solid tumors. Examples of methylating anticancer drugs are procarbazine (PCB, PCZ, N-methyl-hydrazine, Natulan®, Matulane®), dacarbazine (DIC, imidazole carboxamide, dimethyl-triazeno-imidazole-carboxamide, DTIC®-Dome), streptozotocin (STZ, NSC 85998, Zanosar®) and temozolomide (TMZ, SCHS2.365, NSC 362856, Temodal®, Temodar®). PCB and DTIC require metabolic activation to form their DNA-reactive metabolite [5], [6] while STZ alkylates DNA without metabolic activation [7]. TMZ undergoes spontaneous hydrolysis at physiological pH to generate the active metabolite, 5-(3-methyl triazen-1-yl)imidazole-4-carboxamide (MTIC) [8] (Fig. 1). The O6-chloroethylating agents, which include carmustine (BCNU, BiCNU®), lomustine (CCNU, CeeNU®), nimustine (ACNU) and fotemustine (Muphoran®) are used in the treatment of glioblastoma and to a lesser extent in treating malignant melanoma, gastrointestinal and pancreatic cancer, Hodgkin's and non-Hodgkin's lymphoma. Various combinations of methylating and chloroethylating agents together with other anticancer drugs have been used [9], [10], [11] and trials are still ongoing, e.g. in glioma therapy [12].

All of the above-mentioned mutagens and chemotherapeutics react with DNA via an SN1 mechanism to form 12 base adducts and phosphotriester (Fig. 2). The SN1 reaction follows a first-order kinetics that is dependent on the formation of an electrophilic carbonium ion, which covalently binds to nucleophilic sites on DNA. Alkyl DNA base adducts have different stabilities. Thus, N3-methyladenine (N3MeA) and N3-methylguanine (N3MeG) are readily hydrolyzed, while other adducts, e.g. N7-methylguanine (N7MeG) are stable for longer times (T1/2 in vitro: 40–80 h) [13]. O6MeG is more stable and persists in DNA in the absence of O6-methylguanine-DNA methyltransferase (MGMT) [14], [15], [16]. N7MeG and N3MeA are the most frequent methyl adducts comprising 80–85% and 8–18% of total alkyl adducts, respectively. However, O6MeG accounting for O.3 (for methyl methanesulfonate) up to 8% (for methylnitrosourea) of the total DNA methyl adducts is the most critical lesion since it is pre-mutagenic and pre-toxic. Another pre-mutagenic methylation lesion is O4-methylthymine (O4MeT), which is induced at a much lower level (<0.4%) [13]. In this review, agents inducing O6-alkylguanine in DNA are collectively termed O6-alkylating agents (O6AA).

O6MeG and O4MeT are repaired by MGMT (for extensive reviews on MGMT see [17], [18], [19], [20], [21]). If not repaired, O6MeG can give rise to cell death, chromosomal aberrations, mutations and cancer. How much O4MeT contributes to these end points is not precisely known. This review will focus on the important role this DNA repair protein plays in preventing these detrimental endpoints. We will also briefly discuss other alkylation damage defense and processing functions (ABH, BER, MMR) and, finally, the mechanisms behind these endpoints triggered by the MGMT repaired lesions O6MeG and O6-chloroethylguanine.

Section snippets

Single step damage reversal by MGMT

MGMT (also referred to as ATase, AGT, AGAT; E.C. 2.1.1.63) repairs O6-alkylation adducts in a one-step alkyl transfer reaction that transfers the alkyl group from the oxygen in the DNA to a cystein residue in the catalytic pocket of MGMT, thereby restoring DNA and inactivating MGMT. As one MGMT molecule can repair only one alkyl adduct, the cells capacity for removing DNA O6-alkylguanine adducts depends on the total number of MGMT molecules per cell and the rate at which the cell can

One lesion, six endpoints and one defense

Although O6MeG comprises only a small fraction of DNA methyl adducts, it has attracted considerable attention because of its pleiotropic biological effects. O6MeG is responsible for causing point mutations, sister chromatid exchanges (SCEs), chromosomal aberrations, tumor initiation, tumor progression and cell kill. These endpoints, and the role of MGMT in protecting against them, will be discussed below.

Acknowledgements

This work was supported by Deutsche Forschungsgemeinschaft, grants KA724/13-1 and 13-2 and SFB 432/B7. We are grateful to Sankar Mitra and Geoff Margison for helpful comments and proofreading of the manuscript. We acknowledge Julia Hammerling for apoptosis measurements.

References (217)

  • P. Koivisto et al.

    Demethylation of 3-methylthymine in DNA by bacterial and human DNA dioxygenases

    J. Biol. Chem.

    (2004)
  • D.M. Wilson et al.

    The major human abasic endonuclease: formation, consequences and repair of abasic lesions in DNA

    Mutat. Res.

    (2001)
  • D. Chakravarti et al.

    Cloning and expression in Escherichia coli of a human cDNA encoding the DNA repair protein N-methylpurine-DNA glycosylase

    J. Biol. Chem.

    (1991)
  • T. Coquerelle et al.

    Overexpression of N-methylpurine-DNA glycosylase in Chinese hamster ovary cells renders them more sensitive to the production of chromosomal aberrations by methylating agents—a case of imbalanced DNA repair

    Mutat. Res.

    (1995)
  • H. Fung et al.

    A vital role for Ape1/Ref1 protein in repairing spontaneous DNA damage in human cells

    Mol. Cell

    (2005)
  • C.W. Op het Veld et al.

    Methyl methanesulfonate-induced hprt mutation spectra in the Chinese hamster cell line CHO9 and its xrcc1-deficient derivative EM-C11

    Mutat. Res.

    (1998)
  • R.W. Sobol et al.

    Base excision repair intermediates induce p53-independent cytotoxic and genotoxic responses

    J. Biol. Chem.

    (2003)
  • G.E. Kisby et al.

    Role of nucleotide- and base-excision repair in genotoxin-induced neuronal cell death

    DNA Repair (Amsterdam)

    (2004)
  • R. Prasad et al.

    DNA polymerase beta-mediated long patch base excision repair. Poly(ADP-ribose)polymerase-1 stimulates strand displacement DNA synthesis

    J. Biol. Chem.

    (2001)
  • K.W. Caldecott

    XRCC1 and DNA strand break repair

    DNA Repair (Amsterdam)

    (2003)
  • Z.P. Chen et al.

    Relationship between O6-methylguanine-DNA methyltransferase levels and clinical response induced by chloroethylnitrosourea therapy in glioma patients

    Can. J. Neurol. Sci.

    (1999)
  • T. Bessho

    Induction of DNA replication-mediated double strand breaks by psoralen DNA interstrand cross-links

    J. Biol. Chem.

    (2003)
  • W.P. Roos et al.

    DNA damage-induced cell death by apoptosis

    Trends Mol. Med.

    (2006)
  • J. Jiricny

    MutLalpha: at the cutting edge of mismatch repair

    Cell

    (2006)
  • S. Gradia et al.

    hMSH2-hMSH6 forms a hydrolysis-independent sliding clamp on mismatched DNA

    Mol. Cell

    (1999)
  • L.J. Blackwell et al.

    Distinct MutS DNA-binding modes that are differentially modulated by ATP binding and hydrolysis

    J. Biol. Chem.

    (2001)
  • J. Genschel et al.

    Human exonuclease I is required for 5′ and 3′ mismatch repair

    J. Biol. Chem.

    (2002)
  • M.J. Longley et al.

    DNA polymerase delta is required for human mismatch repair in vitro

    J. Biol. Chem.

    (1997)
  • R. Fishel et al.

    The human mutator gene homolog MSH2 and its association with hereditary non-polyposis colon cancer

    Cell

    (1993)
  • F.S. Leach et al.

    Mutations of a mutS homolog in hereditary non-polyposis colorectal cancer

    Cell

    (1993)
  • H. Bartsch et al.

    Relevance of nitrosamines to human cancer

    Carcinogenesis

    (1984)
  • L.J. Marnett et al.

    Endogenous DNA adducts: potential and paradox

    Chem. Res. Toxicol.

    (1993)
  • L. Goria-Gatti et al.

    In vitro and in vivo evidence for the formation of methyl radical from procarbazine: a spin-trapping study

    Carcinogenesis

    (1992)
  • J.M. Reid et al.

    Metabolic activation of dacarbazine by human cytochromes P450: the role of CYP1A1, CYP1A2, and CYP2E1

    Clin. Cancer Res.

    (1999)
  • S. Ostermann et al.

    Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients

    Clin. Cancer Res.

    (2004)
  • J.J. Raizer et al.

    Phase 1 study of 28-day, low-dose temozolomide and BCNU in the treatment of malignant gliomas after radiation therapy

    Neurooncology

    (2004)
  • A. Silvani et al.

    Phase II trial of cisplatin plus temozolomide, in recurrent and progressive malignant glioma patients

    J. Neurooncol.

    (2004)
  • U. Herrlinger et al.

    Phase II trial of lomustine plus temozolomide chemotherapy in addition to radiotherapy in newly diagnosed glioblastoma: UKT-03

    J. Clin. Oncol.

    (2006)
  • R. Goth et al.

    Persistence of O6-ethylguanine in rat-brain DNA: correlation with nervous system-specific carcinogenesis by ethylnitrosourea

    Proc. Natl. Acad. Sci. U.S.A.

    (1974)
  • R. Goth-Goldstein

    Inability of Chinese hamster ovary cells to excise O6-alkylguanine

    Cancer Res.

    (1980)
  • G.P. Margison et al.

    O6-alkylguanine-DNA alkyltransferase: role in carcinogenesis and chemotherapy

    Bioessays

    (2002)
  • R.S. McElhinney et al.

    O6-alkylguanine-DNA alkyltransferase inactivation in cancer chemotherapy

    Mini Rev. Med. Chem.

    (2003)
  • S.L. Gerson

    MGMT: its role in cancer aetiology and cancer therapeutics

    Nat. Rev. Cancer

    (2004)
  • K.S. Srivenugopal et al.

    Ubiquitination-dependent proteolysis of O6-methylguanine-DNA methyltransferase in human and murine tumor cells following inactivation with O6-benzylguanine or 1,3-bis(2-chloroethyl)-1-nitrosourea

    Biochemistry

    (1996)
  • M. Xu-Welliver et al.

    Degradation of the alkylated form of the DNA repair protein, O(6)-alkylguanine-DNA alkyltransferase

    Carcinogenesis

    (2002)
  • S.R. Mullapudi et al.

    DNA repair protein O6-alkylguanine-DNA alkyltransferase is phosphorylated by two distinct and novel protein kinases in human brain tumour cells

    Biochem. J.

    (2000)
  • K.S. Srivenugopal et al.

    Protein phosphorylation is a regulatory mechanism for O6-alkylguanine-DNA alkyltransferase in human brain tumor cells

    Cancer Res.

    (2000)
  • A. Lim et al.

    The nuclear targeting and nuclear retention properties of a human DNA repair protein O6-methylguanine-DNA methyltransferase are both required for its nuclear localization: the possible implications

    EMBO J.

    (1996)
  • R.S. Day et al.

    Defective repair of alkylated DNA by human tumour and SV40-transformed human cell strains

    Nature

    (1980)
  • D.B. Yarosh et al.

    Repair of O6-methylguanine in DNA by demethylation is lacking in Mer-human tumor cell strains

    Carcinogenesis

    (1983)
  • Cited by (526)

    • Precision medicine in gastroenteropancreatic neuroendocrine neoplasms: Where are we in 2023?

      2023, Best Practice and Research: Clinical Endocrinology and Metabolism
    View all citing articles on Scopus
    View full text