Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


The human alkylation B (AlkB) homologs, ALKBH2 and ALKBH3, respond to methylation damage to maintain genomic integrity and cellular viability. Both ALKBH2 and ALKBH3 are direct reversal repair enzymes that remove 1-methyladenine (1meA) and 3-methylcytosine (3meC) lesions commonly generated by alkylating chemotherapeutic agents. Thus, the existence of deficiencies in ALKBH proteins can be exploited in synergy with chemotherapy. In this study, we investigated possible interactions between ALKBH2 and ALKBH3 with other proteins that could alter damage response and discovered an interaction with the mismatch repair (MMR) system. To test whether the lack of active MMR impacts ALKBH2 and/or ALKBH3 response to methylating agents, we generated cells deficient in ALKBH2, ALKBH3, or both in addition to Mlh homolog 1 (MLH1), another MMR protein. We found that MLH1koALKBH3ko cells showed enhanced resistance toward SN1- and SN2-type methylating agents, whereas MLH1koALKBH2ko cells were only resistant to SN1-type methylating agents. Concomitant loss of ALKBH2 and ALKBH3 (ALKBH2ko3ko) rendered cells sensitive to SN1- and SN2-agents, but the additional loss of MLH1 enhanced resistance to both types of damage. We also showed that ALKBH2ko3ko cells have an ATR-dependent arrest at the G2/M checkpoint, increased apoptotic signaling, and replication fork stress in response to methylation. However, these responses were not observed with the loss of functional MLH1 in MLH1koALKBH2ko3ko cells. Finally, in MLH1koALKBH2ko3ko cells, we observed elevated mutant frequency in untreated and temozolomide treated cells. These results suggest that obtaining a more accurate prognosis of chemotherapeutic outcome requires information on the functionality of ALKBH2, ALKBH3, and MLH1.

Free full text 


Logo of jbcThe Journal of Biological Chemistry
J Biol Chem. 2024 Aug; 300(8): 107492.
Published online 2024 Jun 24. https://doi.org/10.1016/j.jbc.2024.107492
PMCID: PMC11326903
PMID: 38925328

Lack of mismatch repair enhances resistance to methylating agents for cells deficient in oxidative demethylation

Associated Data

Supplementary Materials
Data Availability Statement

Abstract

The human alkylation B (AlkB) homologs, ALKBH2 and ALKBH3, respond to methylation damage to maintain genomic integrity and cellular viability. Both ALKBH2 and ALKBH3 are direct reversal repair enzymes that remove 1-methyladenine (1meA) and 3-methylcytosine (3meC) lesions commonly generated by alkylating chemotherapeutic agents. Thus, the existence of deficiencies in ALKBH proteins can be exploited in synergy with chemotherapy. In this study, we investigated possible interactions between ALKBH2 and ALKBH3 with other proteins that could alter damage response and discovered an interaction with the mismatch repair (MMR) system. To test whether the lack of active MMR impacts ALKBH2 and/or ALKBH3 response to methylating agents, we generated cells deficient in ALKBH2, ALKBH3, or both in addition to Mlh homolog 1 (MLH1), another MMR protein. We found that MLH1koALKBH3ko cells showed enhanced resistance toward SN1- and SN2-type methylating agents, whereas MLH1koALKBH2ko cells were only resistant to SN1-type methylating agents. Concomitant loss of ALKBH2 and ALKBH3 (ALKBH2ko3ko) rendered cells sensitive to SN1- and SN2-agents, but the additional loss of MLH1 enhanced resistance to both types of damage. We also showed that ALKBH2ko3ko cells have an ATR-dependent arrest at the G2/M checkpoint, increased apoptotic signaling, and replication fork stress in response to methylation. However, these responses were not observed with the loss of functional MLH1 in MLH1koALKBH2ko3ko cells. Finally, in MLH1koALKBH2ko3ko cells, we observed elevated mutant frequency in untreated and temozolomide treated cells. These results suggest that obtaining a more accurate prognosis of chemotherapeutic outcome requires information on the functionality of ALKBH2, ALKBH3, and MLH1.

Keywords: methylating agents, mismatch repair, AlkB, demethylation, MLH1

DNA damage induced by methylating agents produces a range of DNA lesions that pose a considerable threat to human health. Nevertheless, methylating agents, such as temozolomide (TMZ), are commonly used during chemotherapy, with the goal of killing rapidly replicating cancer cells. Methylating agents induce cytotoxicity by generating DNA lesions that actively block DNA replication, thereby increasing genomic instability in the form of stalled/collapsed replication forks and single-strand (ss) and double-strand (ds)breaks. Methylating agents belong to two classes, SN1 or SN2. SN1 agents involve a first-order reaction at a nucleophilic site with formation of a carbonium ion while SN2 are second order reactions where a transition complex is formed that is dependent on the concentration of both reactants (1). Treatment of cells with SN1- or SN2-methylating agents gives rise to 7-methylguanine (7meG), 3-methyladenine (3meA), mgmt lesions in DNA (2). N-methylpurine-DNA glycosylase (MPG, also referred to as AAG, APDG, and ANPG) efficiently removes the major adducts, 7-methylguanine and 3-methyladenine, resulting in the formation of abasic sites that are processed by downstream base excision repair proteins (3, 4). However, minor damage sites, 1-methyladenine (1meA) and 3-methylcytosine (3meC), although produced at lower amounts, can have significant cytotoxic and mutagenic responses (2). In humans, the major repair proteins involved in repairing 1meA and 3meC are the direct reversal repair (DRR) proteins, alkylation B homologs 2 and 3 (ALKBH2 and ALKBH3).

The AlkB family of dioxygenases directly remove DNA damage via an α-ketoglutarate-dependent oxidative demethylation reaction. In Escherichia coli, AlkB repairs 1meA and 3meC adducts present on ssDNA, dsDNA, and RNA (5, 6). E. coli AlkB mutants show increased sensitivity to methylating agents, and accumulate methyl adducts that correlate with elevated mutant frequency (7, 8). In mammalian cells, there are nine different AlkB homologs: ALKBH1-8 and fat mass and obesity associated gene. However, only ALKBH2 and ALKBH3 appear to have similar repair activities to E. coli AlkB (9, 10). ALKBH2 is primarily located in the nucleus, where it colocalizes with proliferating cell nuclear antigen during S-phase and shows a strong affinity for repairing damage on dsDNA (10, 11, 12). In contrast, ALKBH3 is in both the cytoplasm and nucleus, where it is associated with activating signal cointegrator complex 3 to facilitate repair of damage on RNA and ssDNA (10, 13, 14). Methyl lesions that are repaired by AlkB, ALKBH2, and ALKBH3 block DNA replication, transcription, and interfere with base-pairing (7, 15, 16). Mouse embryonic fibroblast (MEF) cell lines derived from Alkbh2-or Alkbh3-null mice are sensitive to SN2-type reagent methyl methanesulfonate (MMS) treatment (17, 18). Moreover, in the absence of exposure to methylating agents, Alkbh2-null mice accumulate 1meA lesions in their liver DNA, due to endogenous methylation (18). The 1meA and 3meC lesions that are removed by the ALKBH enzymes are bypassed by translesion synthesis DNA polymerases, which result in mutations (15, 16). Alkbh2-deficient MEFs also exhibit an increase in mutant frequency after treatment with MMS (17). Although these studies highlight the biological significance of ALKBH2 and ALKBH3, more research is required to further understand how these enzymes protect against the toxicity and mutagenicity of methylating agents.

The mismatch repair (MMR) pathway is tightly conserved from bacteria to humans. MMR is a postreplication mechanism that is responsible for repairing errors generated during DNA synthesis. Recognition of base-base mismatches is initiated by one of the MutS complexes, MutSα homologs (MSH2-MSH6) or MutSβ (MSH2-MSH3). The MutS complex then recruits MutLα (MLH1-PMS2). Removal of the strand is carried out by Exo1, followed by DNA synthesis by POLδ, and sealing of the backbone by LIG1. In addition to the repair of normal base mismatches, MMR also recognizes numerous damaged bases (19, 20, 21, 22, 23, 24). Increasing evidence suggests that MMR machinery works within the same pathway, or has overlapping functions, for lesions normally repaired by base excision repair, nucleotide excision repair, double-strand break (DSB) repair, and DRR (22, 25, 26, 27, 28, 29, 30, 31). Specifically for methylation damage, MMR recognizes O6-methylguanine (O6meG):T and O6meG:C mismatches that form in O6-methylguanine-DNA methyltransferase (MGMT)-deficient cells and induces cytotoxicity in an ATR-dependent manner (2, 32, 33). The simultaneous elimination of MGMT and MMR results in cells that have increased resistance to methylating agents (based on survival), and also have increased mutant frequency (33, 34, 35, 36, 37). This resistance can counteract the effects of chemotherapeutic treatments (38, 39, 40, 41). However, there are currently no studies that have investigated the interaction between MMR and ALKBH2 and ALKBH3.

In this study, we address the hypothesis that deficiencies in ALKBH2, ALKBH3, or both, along with loss of MLH1 leads to cells that are at least as resistant as WT cells to methylating agent damage. We found that there is an interaction between the MMR-machinery and ALKBH2 in response to treatment with MMS. To explore that interaction further, we produced cells with targeted deletions for ALKBH2, ALKBH3, or both, along with loss of MLH1, and show that there is resistance to treatment with TMZ. Moreover, we describe an MLH1-dependent activation of ATR that leads to a G2/M-cell cycle arrest in cells that are doubly deficient in ALKBH2 and ALKBH3. Also, we found that stress at the replication fork is impeded after the loss of functional MLH1 and ALKBH2 and ALKBH3 (MLH1koALKBH2ko3ko). In addition, our data show that the triple mutant cells have increased mutant frequencies after TMZ treatment, which is consistent with these findings having significant clinical implications.

Results

ALKBH2, an oxidative demethylase, interacts with MSH2, an MMR protein

ALKBH2 protects against DNA damage and mutation, and although it is a DRR protein, we reasoned that there are other proteins it interacts with to repair DNA lesions more efficiently. To identify proteins that interact with ALKBH2, we immunoprecipitated proteins from HEK293T cells using a purified polyclonal ALKBH2 antibody. Proteins were separated using SDS-PAGE electrophoresis and subjected excised bands to trypsinization, followed by analysis using mass spectrometry (MS). Numerous DNA repair proteins interact with ALKBH2, including the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and meiotic recombination 11 homolog (MRE11), both involved in DSB repair (Table 1). Structure-specific recognition protein 1 (SSRP1) is a subunit that along with suppressor of Ty16 (SPT16), form the histone chaperone, which facilitates chromatin transcription elongation. Facilitates chromatin transcription plays important roles in DNA replication and repair by remodeling chromatin (42, 43, 44). In addition to DNA repair proteins, numerous cell cycle proteins bound to ALKBH2, including the protein phosphatase 1 catalytic subunit A and mitogen-activated protein kinase 1 (MAPK1). We chose to focus on the MutS homolog 2 (MSH2) that is part of the MMR system, because MGMT, another DRR protein, also interacts with MSH2 (45). We confirmed the interaction of MSH2 with ALKBH2 using immunoprecipitation and Western blot (Fig. S1D). Similar MS experiments with ALKBH3 did not show an interaction with MSH2 or other MMR complex proteins (Table S3). A complete list of ALKBH2-interacting proteins can be found in the supplemental data (Fig. S1 and Table S2).

Table 1

Selected ALKBH2 interacting proteins from mass spectrometric trypsin peptides

DNA replication and repair
Protein nameProteinScore% Sequence Covered
DNA-dependent protein kinase catalytic subunitDNA-PKcs9.971.8 (72)
Meiotic recombination 11 homologMRE1116.0415.7 (81)
MutS homolog 2MSH24.184.6 (37)
Structure specific recognition protein 1SSRP14.435.8 (37)
Minichromosome maintenance complex component 3MCM332.0717.5 (141)
Minichromosome maintenance complex component 5MCM536.5821.1 (146)

Cell Cycle
Protein nameProteinScore% Sequence covered
CDK5 regulatory subunit-associated protein 2CDK5AP210.817.4 (95)
Casein kinase II subunit alphaCSNK2A113.2214.5 (56)
Mitogen-activated protein kinase 1MAPK14.969.8 (34)
Protein phosphatase 1 catalytic subunit αPPP1A9.6913.6 (42)
Cell division cycle 5CDC516.0212.2 (98)

% sequence covered is the number of peptides matched in parentheses.

Targeted deletions in ALKBH2, ALKBH3, and MLH1 DNA repair genes

The association of MSH2 and ALKBH2 was intriguing considering the interaction between MGMT and MSH2 (45). Although MSH2 functions at the recognition phase of MMR, elimination of Mlh1 at a later stage of MMR resulted in resistance and increased mutations in MEFs (46). The resistance of cells deficient in MGMT and MLH1 showed that MSH2 alone was not responsible for conveying resistance, but rather that a functional MMR system is necessary for sensitivity to methylation damage (33). Therefore, we targeted MLH1 to investigate the interaction between the ALKBH DRR proteins and the MMR pathway. We introduced targeted deletions that eliminated the active site of the protein in ALKBH2 and/or ALKBH3, both of which protect against methylation damage (Figs. 1A and S2) (9, 17). We also targeted MLH1 to explore whether an intact MMR system is critical for the response to methylation damage. To study the link between DRR and MMR, we identified KOs in clones with ALKBH2 or ALKBH3, or both (Fig. 1B). We also generated MLH1 KO cells with mutations in ALKBH2, ALKBH3 or both (Fig. 1C). To evaluate the presence of DRR protein MGMT, we used Western blots to show that MGMT remained present in all the targeted cell lines (Fig. S3).

An external file that holds a picture, illustration, etc.
Object name is gr1.jpg

Targeting and KO validation of MLH1, ALKBH2, and ALKBH3. A, schematic illustrating the target regions ALKBH2, ALKBH3, and MLH1. Indicated regions were cleaved using a dual single-guide RNA CRISPR/Cas9 targeting scheme. B and C, confirmation of MLH1, ALKBH2, and ALKBH3 KOs through immunoblotting. Fifty micrograms of whole cell extracts was subjected to immunoblotting to detect MLH1, ALKBH2, and ALKBH3 expression. β-actin is the loading control. Molecular weights are indicated on the left of the panels. The same β-actin bands are found in Fig. S3. ALKBH, alkylation B homolog; MLH1, Mlh homolog 1.

Loss of ALKBH2 and/or ALKBH3 DNA repair enzymes in cells results in enhanced sensitivity to MMS treatment

We sought to identify the role of the ALKBH DNA repair proteins in mediating cell viability in response to methylation damage. We hypothesized that ALKBH2 and/or ALKBH3 are/is required for inducing cellular sensitivity toward SN2-methylating agents. Because SN2-methylating agents yield a high percentage of 1meA and 3meC lesions relative to SN1-agents, we began by investigating the response to this class of damaging agent (47). To address this hypothesis, we determined cell survival by treating KO cells with increasing concentrations of MMS (an SN2 methylating agent) and staining with crystal violet at 3 days post treatment. We found that a loss of ALKBH2 and/or ALKBH3 results in an overall enhanced sensitivity to MMS compared to WT controls (Fig. 2). Specifically, both ALKBH2ko and ALKBH3ko cells were more sensitive to MMS than WT controls, but only above 500 μM MMS (Fig. 2, A, B, D, and E). At 1.0 mM MMS, both ALKBH2ko and ALKBH3ko cells are twice as sensitive to MMS compared to parental cells (Fig. 2E). These findings are consistent with previous findings, which found that loss of either ALKBH2 or ALKBH3 renders cells sensitive to methylation damage with MMS (17, 18, 48). Simultaneous loss of both ALKBH2 and ALKBH3 (ALKBH2ko3ko) results in enhanced sensitivity to MMS (Fig. 2C). At 0.4 mM MMS, there was a statistically significant decrease in survival in ALKBH2ko3ko cells, almost 3-fold, relative to parental cells (Fig. 2D). Calculation of the IC50 confirmed enhanced sensitivity to MMS in ALKBH2ko, ALKBH3ko, and ALKBH2ko3ko cells (Table S1). These findings indicate that ALKBH2 and ALKBH3 have compensatory and/or overlapping activities for substrates when present; therefore, loss of both repair proteins exacerbates methylation damage.

An external file that holds a picture, illustration, etc.
Object name is gr2.jpg

Cell Survival in HAP1-knockout cells after treatment with MMS. Cell survival of ALKBH2ko, ALKBH3ko, MLH1koALKBH2ko, MLH1koALKBH3ko, and MLH1koALKBH2ko3ko cells was determined after treatment with SN2 methylating agent, MMS. Cells were treated with increasing concentrations of MMS for 1 h, then incubated in fresh-medium. Three days after treatment, cells were fixed with neutral buffered formalin, followed by staining with 0.1% crystal violet solution, and the dye was resolubilized with 10% acetic acid. Relative survival was determined by setting the untreated controls as the baseline. Survival of (A) ALKBH2koversus MLH1koALKBH2ko, (B) ALKBH3koversus MLH1koALKBH3ko, and (C) ALKBH2ko3koversus MLH1koALKBH2ko3ko cells was determined. Survival of all cells after treatment with (D) 0.4 mM or (E) 1.0 mM MMS was analyzed using crystal violet staining. Statistical analysis was carried out using an ordinary one-way ANOVA test. All experiments were performed in triplicate (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ALKBH, alkylation B homolog; MMS, methyl methanesulfonate; MLH1, Mlh homolog 1.

Simultaneous loss of MLH1, ALKBH2, and ALKBH3 leads to resistance to MMS treatments at low concentrations

Next, we sought to identify whether the interaction with the MMR system that was previously observed (Table 1) with ALKBH2 is important for establishing a response to methylation damage. To address this, we used the MLH1ko ALKBH2ko cell line to determine cell survival after MMS treatment. Also, because ALKBH2 and ALKBH3 may have compensatory roles, we investigated a potential role for MMR in cells deficient in ALKBH2, ALKBH3, or both. We found that loss of MLH1 in ALKBH2ko cells (MLH1koALKBH2ko) resulted in enhanced sensitivity toward MMS, specifically at 1.0 mM MMS (Fig. 2, A and E). In contrast, MLH1koALKBH3ko and MLH1koALKBH2ko3ko cells demonstrated resistance to MMS treatments compared to their MLH1-expressing controls (Fig. 2, B and C). At 0.4 mM, MLH1koALKBH2ko3ko cells showed a statistically significant increase in survival as compared to ALKBH2ko3ko cells (Fig. 2D). At 1.0 mM MMS, both MLH1-expressing and MLH1-deficient ALKBH2ko3ko cells show the same sensitivity (Fig. 2E). The IC50 for ALKBH2ko3ko and MLH1koALKBH2ko3ko cells were 387 and 449 μM MMS, respectively (Table S1). In contrast, at 1.0 mM MMS, MLH1koALKBH3ko cells were resistant to treatment as compared to MLH1-expressing controls (Fig. 2E). The IC50 for ALKBH3ko and MLH1koALKBH3ko cells were 469 and 509 μM MMS, respectively (Table S1). These results are surprising because a direct interaction between an MMR protein and ALKBH3 was not observed. In conclusion, these findings indicate that functional MLH1 is required for sensitivity at low MMS concentration in the absence of ALKBH3, but not ALKBH2.

Loss of ALKBH2 and/or ALKBH3 renders cells sensitive to TMZ

Next, we sought to determine the survival of the previously established cell lines after treatment with an SN1-methylating agent. In contrast to SN2-agents, SN1-agents have a different distribution of lesions upon reacting with DNA, and as they are widely used in a clinical setting, understanding the cellular response to these agents has relevance for treatment. We hypothesized that loss of either ALKBH2 or ALKBH3 or both results in enhanced sensitivity to SN1-methylation agents such as TMZ. To address this hypothesis, ALKBH2ko, ALKBH3ko, and ALKBH2ko3ko cells were treated with TMZ, then allowed to recover for 3 days, and cell survival was analyzed using crystal violet staining. We found that cell survival of ALKBH2ko and ALKBH3ko cells remained relatively unchanged when cells were treated within a range of 0 to 500 μM of TMZ (Fig. 3, A and B). The IC50 of ALKBH2ko and ALKBH3ko cells were 446.0 and 452.9 μM TMZ, respectively (Table S1). However, ALKBH2ko3ko cells were extremely sensitive to treatment with TMZ (Fig. 3C). At 500 μM TMZ, ALKBH2ko and ALKBH3ko cells remained viable, but ALKBH2ko3ko cells showed extreme sensitivity (Fig. 3D). At 2.0 mM TMZ, all three cell lines were hypersensitive to treatment (Fig. 3E). These results were recapitulated using a colony formation assay in two independent cell lines (Figs. 3F and S4B). ALKBH2ko3ko had an IC50 of 162 μM TMZ, as compared to >500 μM TMZ, which was extrapolated from the survival curve (Table S1). Taken together, the ALKBH2 and ALKBH3 are both important in repair, not only for MMS-induced damage (SN2), but also for repair of TMZ-induced damage (SN1) at high concentrations.

An external file that holds a picture, illustration, etc.
Object name is gr3.jpg

Cell Survival and Colony Formation in HAP1-KO cells after treatment with TMZ. Cell survival of ALKBH2ko, ALKBH3ko, MLH1koALKBH2ko, MLH1koALKBH3ko, and MLH1koALKBH2ko3ko cells was determined after treatment with the SN1 methylating agent, TMZ, as previously stated. The survival of (A) ALKBH2ko versus MLH1koALKBH2ko, (B) ALKBH3koversus MLH1koALKBH3ko, and (C) ALKBH2ko3koversus MLH1koALKBH2ko3ko cells was determined. Survival of all cells after treatment with (D) 500 μM or (E) 2.0 mM TMZ was analyzed using crystal violet staining. F, a colony formation assay was carried out on ALKBH2ko3ko and MLH1koALKBH2ko3ko cell lines 7 days after treatment with TMZ. Statistical analysis was carried out using an ordinary one-way ANOVA test. All experiments were performed in triplicate (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ALKBH, alkylation B homolog; MLH1, Mlh homolog 1; TMZ, temozolomide.

Loss of MMR in single and double deficient ALKBH2 and/or ALKBH3 cells leads to resistance to TMZ

The results with MMS indicated a potential role between MLH1 and the ALKBH2 and ALKBH3 proteins (Fig. 2). Therefore, we hypothesized that a similar role would be observed after treatment with an SN1-methylating agent. To address that hypothesis, cell survival to TMZ was determined, as previously mentioned using crystal violet staining. We found that loss of MLH1 in either ALKBH2ko (MLH1koALKBH2ko) or ALKBH3ko (MLH1koALKBH3ko) cells did not result in reduced cell viability at 500 μM TMZ (Fig. 3, A, B, and D). However, treatment with 2.0 mM TMZ led to resistance in MLH1koALKBH2ko and MLH1koALKBH3ko cells that was statistically significant when compared to their MLH1-expressing controls (Fig. 3E). The IC50 of MLH1koALKBH2ko and MLH1koALKBH3ko cells were >500 μM, respectively, which was an increase from the ALKBH2ko and ALKBH3ko cells that were previously described (Table S1). Also, we found that the MLH1koALKBH2ko3ko cells showed enhanced resistance to TMZ at 500 μM and 2.0 mM TMZ (Fig. 3, CE). MLH1koALKBH2ko3ko survival showed a cumulative effect in survival when compared against MLH1koALKBH2ko and MLH1koALKBH3ko cells (Fig. 3E). Moreover, the IC50 of MLH1koALKBH2ko3ko cells increased to >500 μM TMZ, as compared to 162 μM in ALKBH2ko3ko cells (Table S1). These results were recapitulated using another independent MLH1koALKBH2ko3ko cell line (Figs. 3F and S4B). Overall, the findings obtained from the MMS and TMZ treatments highlight a potential role for MLH1 in inducing cell death after recognition of methyl lesions that are repaired by ALKBH2 and ALKBH3 for both SN1 and SN2 methylating agents.

MMR-induced replication arrest in the first cell cycle is MLH1-dependent

Lesion recognition by the MMR pathway can result in an MMR-dependent cell cycle arrest in the second G2-phase after methylation damage (32). Based on these findings and the cell survival results, we sought to determine if the resistance to TMZ observed in MLH1koALKBH2ko3ko cells was due to an inability to arrest at the G2 phase. Actively replicating cells were treated with 100 μM TMZ for 1 h, and replication kinetics were determined by pulse-labeling cells with BrdU, and stained with propidium iodide at 24-, 48-, and 72-h post treatment followed by fluorescent-activated cell sorting (FACS) analysis. If direct damage by TMZ is responsible for sensitivity to damage during the first 24 h, then, we expected a significant decrease of cells present in the G2/M-phase immediately following treatment. FACS analysis revealed that the progression of ALKBH2ko3ko cells, although limited, still occurred at 24 h after TMZ treatment, resulting in an accumulation of cells in G2-phase (Fig. 4, A and B). Using BrdU-incorporation, we determined that 27% and 41% of cells were present at the S- and G2/M-phase 24 h after TMZ treatment, respectively (Fig. 4B). These results indicate that cells continued through DNA replication immediately following TMZ treatment. However, at 48 h and 72 h after TMZ treatment, ALKBH2ko3ko cells are completely arrested at the G2-phase (Fig. 4A). However, at 48 and 72 h post treatment, 77.9% and 66.2% of ALKBH2ko3ko cells were arrested at the G2/M-phase, respectively (Fig. 4B).

An external file that holds a picture, illustration, etc.
Object name is gr4.jpg

Replication kinetics of HAP1 cells after 100 μM TMZ treatment. FACS analysis of HAP1 cell cultures with 100 μM TMZ carried out at 24 to 72 h post treatment. The indicated KO cells were treated with 100 μM of TMZ for 1 h, then pulse labeled with 10 μM BrdU for 30 min before harvesting cells at the indicated time points. Cells were fixed with 70% ethanol and stained with PI before FACS analysis. A, FACS analysis carried out 24 to 72 post treatment in HAP1, MLH1ko, ALKBH2ko3ko, and MLH1koALKBH2ko3ko cells. B, cell distribution % of KO cells was collected by quantifying BrdU-labeling at 24, 48, and 72 h post TMZ treatment. Over 50,000 events were collected using the BD Accuri C6 Flow Cytometer. ALKBH, alkylation B homolog; FACS, fluorescent-activated cell sorting; MLH1, Mlh homolog 1; TMZ, temozolomide.

In contrast, MLH1koALKBH2ko3ko cells showed a slight attenuation of progression through replication at 24 h post-TMZ treatment. (Fig. 4A). Using BrdU-incorporation to determine replication kinetics, we found that 29.9% of MLH1koALKBH2ko3ko cells were at the G2/M-phase at 24 h post-TMZ as compared to 20.4% of untreated controls (Fig. 4B). However, replication kinetics returned to levels comparable to parental and MLH1ko controls at 48-h and 72-h post TMZ treatment (Fig. 4, A and B). Thus, these results indicate that a TMZ-induced checkpoint arrest observed in ALKBH2ko3ko cells is dependent on a functional MMR pathway.

G2-arrest is dependent on ATR activation after TMZ treatment

Recognition of drug-induced DNA adducts by the MMR pathway is followed by recruitment of ATM and/or ATR kinases, which activates a DNA damage response (DDR) that cause cell cycle arrest or induction of apoptosis (49, 50). We hypothesized that the G2/M-arrest observed in ALKBH2ko3ko cells was due to an MLH1-dependent activation of a DDR. To address this hypothesis, we sought to identify the DDR kinase that is required for cell cycle arrest at the G2/M-phase in response to TMZ. To determine this, cells were treated with TMZ, then cultured in complete media containing an ATR-inhibitor (ATRi, VE-821) or an ATM-inhibitor (ATMi, KU-55933) for the last 18 h before pulse-labeling with BrdU, staining with propidium iodide, and harvesting. The proposed protocol is outlined in Figure 5A (Fig. S5A), previously used to study the role of MMR and MGMT in replication arrest (32). We did not observe any significant changes in cell cycle distribution and replication kinetics under the presence of ATMi, KU-55933 (Fig. S5, B and C). In contrast, using the ATRi, VE-821, we found that ALKBH2ko3ko cells were unable to enter the first G2/M phase of the cell cycle at 24 h after TMZ treatment (Fig. 5B). At 24 h post-TMZ, we found that 84.6% of ALKBH2ko3ko cells were arrested at the S-phase (Fig. 5C). In comparison 26.9% of ALKBH2ko3ko cells without inhibitors were present in the S-phase at 24 h post TMZ (Fig. 4B). At 48 h, we found that 49.9% of ALKBH2ko3ko cells accumulated at the G2/M phase, but there was still a large population of cells present at the S-phase, 39.2% (Fig. 5C). At 72 h, 68.1% of ALKBH2ko3ko cells were present in the S-phase after 18 h incubation with VE-821 (Fig. 5C). In contrast, MLH1koALKBH2ko3ko cells did not show any overt differences in cell cycle distribution in the presence of an ATRi or ATMi (Figs. 5 and S5). These results indicate that ATR-activation plays a significant role in regulating cell cycle progression in the presence of a functional MMR.

An external file that holds a picture, illustration, etc.
Object name is gr5.jpg

G2/M-arrest in ALKBH2ko3kocells is dependent on ATR activation, not ATM.A, schematic of protocol used to determine the role of ATR inhibition using VE-821. Parental HAP1, MLH1ko, ALKBH2ko3ko, and MLH1koALKBH2ko3ko cells received a treatment of 100 μM TMZ for 1 h. Cells were harvested at 24, 48, and 72 h post treatment in the presence of 5 μM VE-821 during the 18 h. Prior to harvest, cells were pulse labeled with 10 μM BrdU for 30 min. Cells were then fixed with 70% ethanol and stained with PI before FACS analysis. B, FACS analysis carried out 24 to 72 post treatment in HAP1, MLH1ko, ALKBH2ko3ko, and MLH1koALKBH2ko3ko cells. C, cell distribution % of KO cells was collected by quantifying BrdU-labeling at 24, 48, and 72 h post TMZ treatment. Over 50,000 events were collected using the BD Accuri C6 Flow Cytometer. ALKBH, alkylation B homolog; FACS, fluorescent-activated cell sorting; MLH1, Mlh homolog 1; TMZ, temozolomide.

MLH1koALKBH2ko3ko cells have reduced [Latin small letter gamma]H2AX signals and lose apoptotic signaling

DNA lesions that are repaired by ALKBH2 and ALKBH3 are significant cytotoxic lesions due to their ability to block DNA replication (51, 52). These lesions, 1meA and 3meC, if left unrepaired can induce DNA damage in the form of DSBs, or other damage as determined by phosphorylated histone variant H2AX (mgmt) (53). Therefore, we hypothesized that MLH1 is required for inducing DNA damage as indicated by the Ser139 phosphorylation of H2AX in the absence of ALKBH DNA repair enzymes. We addressed this hypothesis by assessing the formation of a phosphorylated histone variant H2AX ([Latin small letter gamma]H2AX), a sensitive marker for DNA damage (54). Cells were treated with 100 μM TMZ for 1 h, and whole-cell protein extracts were subjected to SDS-PAGE. We found a substantial increase in [Latin small letter gamma]H2AX at 48 h in ALKBH2ko3ko cells, but only a small increase (similar to parental cells) in MLH1koALKBH2ko3ko cells (Fig. 6A). As a second measure of DNA damage, we also stained for 53BP1 and then quantified its colocalization with [Latin small letter gamma]-H2AX. We found that MLH1ko significantly increased the levels of 53BP1 and [Latin small letter gamma]-H2AX colocalization. Next, we sought to determine if increased [Latin small letter gamma]H2AX signaling results in changes in apoptotic signaling. We hypothesized that MLH1-dependent activation of a DDR is required for induction of apoptotic signaling. Apoptotic signaling was assessed by examining the cleavage of PARP1, and cells undergoing cellular apoptosis was assessed by measuring Annexin V after treatment to TMZ (55). Immunoblot analysis revealed cleavage of PARP1 at 48 h post TMZ treatment only in ALKBH2ko3ko cells. However, no changes in cleaved PARP1 were observed in parental HAP1, MLH1ko, or MLH1koALKBH2ko3ko cells (Fig. 6B). Apoptosis and cell death were determined by Annexin V-FITC/PI double-staining and quantified by flow cytometry. We found that after 100 μM TMZ treatment, 16.10% and 33.03% of ALKBH2ko3ko cells were undergoing apoptosis at 48 and 72 h, respectively (Fig. 6C). We did not observe an increase in apoptotic cells at 24 h in ALKBH2ko3ko cells. In fact, the number of apoptotic cells of ALKBH2ko3ko cells at 24 h was comparable to parental and MLH1ko controls. Furthermore, we did not observe any change in apoptotic cells in MLH1koALKBH2ko3ko cells at 24, 48, and 72 h post-TMZ treatment (Fig. 6C). Analysis of colocalization of 53BP1 and γ-H2AX showed a significant decrease following TMZ treatment for 24 h in MLH1koALKBH2ko3ko compared to WT (Fig. S6). Therefore, our results support the hypothesis that the presence of functional MMR is fundamentally necessary in cells defective in ALKBH DNA repair proteins for inducing DNA damage in the form of DNA breaks and activating cellular apoptosis in response to methylation damage.

An external file that holds a picture, illustration, etc.
Object name is gr6.jpg

[Latin small letter gamma]H2AX, and apoptotic signaling in HAP1 cells after TMZ treatment. HAP1 cells were treated with 100 μM TMZ for 1 h and collected at the indicated time point. A, Thirty micrograms of whole cell protein extracts were subjected to electrophoresis on a 10 to 15% SDS-PAGE followed by immunoblotting using anti-[Latin small letter gamma]H2AX antibodies. Apoptotic signaling was measured by subjecting 30 μg of whole cell protein extracts to SDS-PAGE followed by immunoblotting using (B) anti-PARP1. β-actin was used as the loading control. C, apoptotic cell death was analyzed in HAP1 cells after TMZ treatment using the Annexin-C/PI staining method. Data represent HAP1 cells stained with PI and Annexin-V conjugated with FITC at the indicated time point after treatment with TMZ. Data represented in this figure represents flow cytometry results of 10,000 events. [Latin small letter gamma]H2AX, phosphorylated histone variant H2AX; TMZ, temozolomide.

After TMZ treatment, MLH1koALKBH2ko3ko cells show reduced replication fork stress as compared to either MLH1ko or ALKBH2ko3ko cells

Because MLH1koALKBH2ko3ko cells did not undergo significant changes in cell cycle progression and [Latin small letter gamma]H2AX signaling post methylation damage, we investigated the effects of MLH1 and the ALKBH2 and ALKBH3 repair protein deficiencies on the replication fork. We used a DNA fiber combing technique to evaluate possible differences in DNA replication that depend on treatment and genetic background. We hypothesized that the most stress at active replication forks would occur in cells that have functional MLH1 and deficient ALKBH2 and ALKBH3. To investigate this hypothesis, HAP1 cells were pulse-labeled with the nucleotide analog IdU for 30 min, followed by a second pulse-label with CldU in combination with 500 μM TMZ or mock treatment with dimethylsulfoxide (DMSO) (Fig. 7A). Using 500 μM TMZ allowed us to determine replication fork kinetics because we show that at this concentration, there was a higher incidence of DNA breaks and apoptotic signaling at 24 h post treatment in ALKBH2ko3ko (Fig. S7). This indicates that substantial DNA damage at this concentration leads to replication stress during the first round of replication. Using DNA fiber analysis, we found that stalled replication forks were observed in MLH1ko or ALKBH2ko3ko cell lines after TMZ treatment. However, no significant changes in fork stalling were observed in MLH1koALKBH2ko3ko cells (Fig. 7B). These results indicate that MLH1, ALKBH2, and ALKBH3 help protect against replication fork stress induced by methylating agents.

An external file that holds a picture, illustration, etc.
Object name is gr7.jpg

MMR, ALKBH2, and ALKBH3 protect against stalled replication forks from methylation damage. DNA fiber combing experiments showing replication fork stability after 500 μM TMZ in HAP1 cells. A, schematic representation of the assay conditions. HAP1 cells were pulse labeled with IdU for 30 min, followed by pulse labeling with CldU for 30 min, in combination with 500 μM TMZ. Fork images show an active fork with IdU and CldU labeling of equal lengths. Stalled forks are observed as reduced CldU length relative to IdU. B, DNA fiber combing experiment shows stalled replication forks in MLH1ko and ALKBH2ko3ko cell lines. CldU and IdU fiber tracts were measured using Zen 2.6 software (Zeiss). The ratio of CldU to IdU tracts lengths is presented. Statistical analysis was carried out using an ordinary one-way ANOVA test. Hundred fibers were counted per group (n = 100). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ALKBH, alkylation B homolog; MMR, mismatch repair; MLH1, Mlh homolog 1; TMZ, temozolomide.

Mutation frequency increases in MLH1koALKBH2ko3ko cells as compared to HAP1 WT cells

The protection of ALKBH2 against the mutagenic effects of 1meA and 3meC in MEFs is established, but the role of the MMR pathway is not known (17). Therefore, we determined the impact of the MLH1 or ALKBH2 and ALKBH3 on mutant frequencies using cells with targeted deletions in those genes, separately or in combination. Cells were treated with 50 μM TMZ, a dose at which 95% of the cells remain viable as indicated by clonal survival assays. To detect mutations, we used a mutation selection assay that monitors Na+/K+-ATPase function. This assay selects for cells that develop resistance to ouabain due to mutations in Na+/K+-ATPase, resulting in colony formation in the presence of this inhibitor (56). Control HAP1 cells produced only a limited number of mutations, whereas MLH1ko cells manifested increased mutant frequencies alone or with TMZ treatment as compared to the HAP1 parental line (Fig. 8). Mutant frequencies obtained in ALKBH2ko3ko cells were almost identical without or with TMZ exposure and similar to the mutant frequency observed for untreated MLH1ko cells (Fig. 8). However, the untreated MLH1koALKBH2ko3ko cells exhibited a 10-fold increase in mutation frequency as compared to HAP1 parental cells and even greater than MLH1ko cells, which are often referred to as having a “mutator phenotype”. However, after exposure to 50 μM TMZ, the MLH1koALKBH2ko3ko cells showed an 80-fold increase in mutant frequency as compared to parental controls. These results support the hypothesis that MLH1 and ALKBH2 and ALKBH3 protect against genetic mutations due to endogenous and exogenous sources of methylation damage. Moreover, loss of MLH1 along with the ALKBH2 and ALKBH3 enzymes cause a large increase in mutant frequency that is synergistic.

An external file that holds a picture, illustration, etc.
Object name is gr8.jpg

Mutant frequency is elevated in MLH1koALKBH2ko3kocells after TMZ treatment. MLH1koALKBH2ko3ko cells show elevated mutation frequency after treatment with TMZ. HAP1 cells were treated with DMSO or 50 μM TMZ for 1 h. Treated cells were expanded for 5 days, followed by treatment of cells in 1 μM of ouabain for 10 days. Ouabain-resistant colonies were fixed and stained, and the mutation frequencies were calculated. Statistical analysis was carried out using a two-way ANOVA. All experiments were performed in triplicates (n = 3). ****p < 0.0001. ALKBH, alkylation B homolog; DMSO, dimethylsulfoxide; MLH1, Mlh homolog 1; TMZ, temozolomide.

Discussion

In this report, we have demonstrated that the elimination of ALKBH2 and ALKBH3 sensitizes cells to methylating agents only in the presence of functional MMR. In cells absent of both ALKBH2 and ALKBH3 DNA repair proteins, but with a functional MMR system, replication is slowed due to methylation damage, consistent with persistent DNA damage inducing stress at the replication fork. Loss of ALKBH2 and/or ALKBH3 along with MMR leads to resistance to treatment with the SN1-agent, TMZ. Moreover, loss of MLH1 in ALKBH3-deficient cells leads to resistance to treatment with MMS and TMZ. Because simultaneous loss of MLH1, ALKBH2, and ALKBH3 manifests the highest resistance to TMZ, we sought to further investigate this effect with an SN1 agent.

Our data show that treatment of cells deficient in both ALKBH2 and ALKBH3 are sensitive to both SN1 and SN2 alkylating agents. We showed that loss of ALKBH2 and ALKBH3 proteins results in a G2/M-arrest in an ATR-dependent manner, elevated DNA breaks, and induction of cellular apoptosis in response to methylation damage. These effects are not observed with concomitant deletion of MLH1. Furthermore, we show that in the absence of methylation damage, the mutant frequency of the parental cell line is extremely low, whereas the mutant frequency of the ALKBH2 and ALKBH3 deficient cell lines and MLH1 deficient cells are similar. However, without TMZ treatment, MLH1koALKBH2ko3ko cells have a mutant frequency that is five-fold that of the ALKBH2ko3ko cells. Moreover, comparing parental cells to MLH1koALKBH2ko3ko, treatment with TMZ results in an 80-fold increase in mutant frequency. Those results are consistent with the higher survival being accompanied by an increase in mutant frequency.

Our findings show that cellular sensitivity to methylation damage in ALKBH2 and ALKBH3 deficient cells is MLH1-dependent, which is comparable to prior studies performed in MGMT deficient cells requiring functional MMR for initiation of cell death (22, 57, 58, 59, 60). The necessity of an intact MMR response in human cells was previously established and is often used as a marker for determining the chemotherapeutic response of certain cancers (33, 61, 62, 63). Moreover, Mgmt−/−Mlh1−/− mouse lung fibroblast cells are resistant to methylation agent damage, with reduced caspase-3 activity after methylating agent treatment (33). Similarly, we noted that there is apoptotic cell death signaling that is MLH1-dependent in ALKBH2ko3ko cells that was measured by cleaved PARP and Annexin/PI staining. Interestingly, this does not occur immediately following damage, but occurs 48 and 72 h post-TMZ treatment. Thus, in the present study, we have extended the influence of MLH1 to ALKBH2 and ALKBH3 in inducing an apoptotic response after methylation damage.

These results show that there is an increase in stalled replication forks in MLH1-deficient and in ALKBH-deficient cells in response to methylation damage. However, TMZ exposure in MLH1koALKBH2ko3ko cells results in no disruption at the replication fork as compared to the TMZ treated parental cell lines. DNA methylation damage inhibits replication elongation (64). Although it is widely accepted that methylation damage blocks DNA polymerases, the number of studies that concentrate on their effect at the replication fork is limited (65). Damage induced by MMS physically blocks actively replicating forks due to high levels of 1meA and 3meC lesions at single-stranded DNA regions (7). Replication block by MMS occurs independently of damage signaling and ss breaks (65). In contrast, SN1-agents, like TMZ, generate 10 times lower levels of 1meA and 3meC lesions on ss and dsDNA (66). Given that replication stress was alleviated in MLH1koALKBH2ko3ko cells, determining the interaction between MMR and 1meA and/or 3meC lesions will allow for a better understanding of the role of MMR on replication stress. Moreover, whether MMR is involved in detecting, and activating a DDR on newly formed 1meA or 3meC lesions also remains to be investigated.

Upon exposure to TMZ, ALKBH2ko3ko cells were arrested at the G2/M phase 48 h post treatment. These results were surprising because arrest usually occurs within a few hours after DNA damage. Therefore, a postdamage molecular mechanism is responsible for inducing arrest. Stojic et. al. found that a G2/M arrest in 293T Lα+ cells was dependent on ATR activation after methylation damage with the SN1-type agent, MNNG (32). The findings presented here show that treatment of ALKBH2ko3ko cells with the ATR inhibitor (VE-821) resulted in an inability to progress through S-phase 48 h post-TMZ treatment. Instead, a vast majority of cells began to undergo apoptosis at 48 and 72 h post treatment. Using the ATMi (KU-55933), no differences in replication kinetics as compared to the ALKBH2ko3ko TMZ-treated cells were detected. Therefore, these results indicate that ATR signaling, not ATM, plays a significant role in preserving genomic integrity during S-phase progression, and G2-arrest. However, this dependence on ATR activation was not present in MLH1koALKBH2ko3ko and MLH1ko cells, thereby indicating a novel role for MMR in activating ATR-signaling in response to 1meA and 3meC damage generated by methylating agents. These results provide evidence that the response to methylation damage in ALKBH2ko3ko cells is dependent on functional MMR to activate a DDR, induce DNA breaks, and activate apoptosis.

The causes of cell death are important but understanding the mechanism for increased resistance for ALKBH- and MLH1-deficient cells could lead to methods that would overcome resistance. Currently, two competing hypotheses describe the role that MMR plays in alkylation damage-induced cytotoxicity with respect to MGMT. The first hypothesis states that in the absence of MGMT, O6meG lesions interfere with base pairing between cytosine, and can pair with thymine during DNA replication. The resulting imperfect base pairing is recognized and processed by MMR proteins during a second round of replication in a futile repair mechanism, which involves the removal of the newly synthesized DNA strand, thereby leaving the O6meG lesion intact. Perpetual processing of O6meG:T pairs induces replication stress in the form of single-stranded DNA breaks, DNA replication blocks, fork collapse, and ultimately DSBs. The second model states that MMR recognition of O6meG:T mismatches activates a DNA damage signaling response by recruiting ATR kinase, which activates a checkpoint response (67). At present, our results provide evidence that favors the futile repair mechanism (32), but the mechanism underlying induction of cell death and the roles of ALKBH2, ALKBH2, and MLH1 should be included in future discussions on this topic.

The persistence of lesions in MLH1koALKBH2ko3ko cells leads to increased mutant frequencies as compared to the parental cell line. In addition to O6meG, methylating agents generate a range of methyl-lesions that can form mispairs and induce mutations if left unrepaired. The DRR proteins, ALKBH2 and ALKBH3, play an essential role in repairing 1meA and 3meC lesion which induce mutations (17). Bypass of 1meA lesions by POLη, POLζ, or POLι/POLδ leads to continued DNA synthesis, and persistence of 1meA within DNA (16). Despite being bypassed by translesion DNA polymerases, replication bypass of 1meA lesions occurs in an error-free manner (16). Moreover, 3meC lesions are also bypassed by translesion DNA polymerases, POLη, POLι, and less efficiently by POLκ (15). Bypass of 3meC leads to misincorporation of dTTP and dATP opposite the lesions (15). These findings are consistent with previous studies in Alkbh2-deficient MEFs which show an increase in C→A and C→T mutations when treated with MMS (17). This suggests that bypass of 3meC in cells occurs through POLη and/or POLι. In this report, we have shown significant increases in the mutant frequencies in MLH1koALKBH2ko3ko cells as compared to the HAP1 background. Moreover, the role between the ALKBH2 and ALKBH3 proteins and MLH1 was not previously reported.

As methylating agents, such as TMZ and dacarbazine, are commonly used for the treatment of glioblastoma, malignant melanoma, and Hodgkin’s and non-Hodgkin’s lymphoma, developing an understanding of the cellular response to methylation damage has significant clinical implications (22, 33, 57, 58, 59, 60). In our study, we observed increased mutant frequency in MLH1koALKBH2ko3ko cells after exposure to TMZ treatment. Unrepaired O6meG lesions in MMR-deficient cells contribute to a mutator phenotype in normal cells that can lead to cancer and that can assist in the formation of chemoresistant cancer cells upon selection (68, 69, 70, 71). The O6meG lesion is considered a highly mutagenic lesion due to its ability to be readily bypassed by translesion DNA polymerases and base pair with thymine (72, 73). However, unlike O6meG, 1meA and 3meC lesions are considered less mutagenic, but have increased mutagenicity owing to their ability to be bypassed during translesion DNA synthesis (7, 8, 15).

These results provide insight into how resistance to methylation damage can arise due to a loss of ALKBH2 and ALKBH3 function. In this study, we generated KOs of both ALKBH proteins to mirror the response that can occur during metabolic inactivation to both proteins. Such inactivation can occur due to genomic mutations in IDH1 and/or IDH2 that frequently occur in gliomas and produce D-2-HG, a competitive inhibitor of αKG. Elevated levels of D-2-HG inhibit ALKBH2 and ALKBH3 and sensitize cells to alkylating agents (53). Mutations in MMR proteins and IDH1 are frequently observed in TMZ resistant gliomas (41). However, the role that IDH mutations play in resistance to methylation damage remains unknown. In this study, we provide a possible explanation for the resistance to methylating agents that could arise from ALKBH deficiency due to D-2-HG coupled with MLH1 or MSH2 silencing.

In conclusion, these results show that loss of ALKBH2 and ALKBH3 in MLH1-deficient cells play a significant role in the development of resistance to methylating agents. In this study, we show that resistance to methylation damage is dependent on ATR signaling, and reduced replication stress. ATR activation arrest cells in the second G2/M phase post damage, potentially due to elevated levels of replication stress, resulting in DNA breaks. Furthermore, inability to activate ATR signaling in MLH1-deficient cells reduced stress at the replication fork, and the persistence of methyl lesions can lead to induced mutation burden. Therefore, our results suggest a novel role for MMR in the recognition of methylation damage that does not include O6meG lesions. Our present studies indicate the importance of understanding the function of DRR, including ALKBH2, ALKBH3, and MGMT along with MMR in evaluating the efficacy of chemotherapeutic methylating agents.

Experimental procedures

Cell culture and drug treatment

HAP1 cells, originally derived from chronic myeloid leukemia cells but transduced using the Yamanaka factors and selected as adherent cells, were purchased from Horizon Discovery (74). The authenticity of cell lines was verified using short tandem repeat profiling. Cells were regularly checked for mycoplasma. Cells were cultured in Iscove’s modified Dulbecco’s medium (IMDM; Gibco) supplemented with 10% fetal bovine serum (FBS; Corning) at 37 °C, 5% CO2. TMZ (TCI America) was dissolved in DMSO at a concentration of 103 mM, and aliquots were stored in −20 °C. MMS (Acros) aliquots were stored in −20 °C. MMS was diluted to 1 mM prior to use. For treatment of exponentially growing cells, TMZ and MMS were added directly to serum-free media. HEK293T cells were purchased from American Type Culture Collection.

Antibodies

ALKBH2 and ALKBH3 human purified proteins were provided to Cocalico Biologicals for inoculation of two rabbits under a general IACUC protocol for antibody production. Sera were collected from the rabbits after 3 months. Antibodies were purified from sera using Montage Antibody Purification Kit (Millipore). The following antibodies were purchased from suppliers: rat anti-BrdU (Novus, BU1/75), mouse anti-BrdU (Becton Dickinson, AB_400326), mouse anti-MLH1 (Santa Cruz Biotechnology, sc-582, epitope is the C terminus), rabbit anti-ALKBH3 (Invitrogen, PA5-52672, epitope is amino acids 14–139), rabbit anti-Caspase 3 (Cell Signaling Technology, SA1E), rabbit anti-PARP1 (Santa Cruz Biotechnology, sc-25780), rabbit anti-[Latin small letter gamma]H2Ax (Cell Signaling Technology, 20E3), mouse anti-β-Actin (Sigma-Aldrich, AC-74), rabbit anti-β-Actin (Sigma-Aldrich, RM112), AlexaFluor 488 chicken anti-rat (Invitrogen, AB_2535873), AlexaFluor 594 rabbit anti-mouse (Invitrogen, AB_2534109), AlexaFluor 488 goat anti-chicken (Invitrogen, AB_2534096), AlexaFluor 594 goat anti-rabbit (Invitrogen, AB_2534079), IRDye-680RD goat anti-rabbit (Li-Cor, AB_2721181), IRDye-800CW goat anti-mouse (Li-Cor, RRID AB_2687825), IRDye-680RD goat anti-mouse (Li-Cor, RRID AB_2651128), and IRDye-800CW goat anti-rabbit (Li-Cor, RRID AB_2651127).

Mass spectrometric analysis of ALKBH2 interacting proteins

HEK293T cells (5 × 150 mm dishes) were exponentially grown in Dulbecco's modified Eagle's medium (DMEM) media containing 10% FBS and lysed in lysis buffer (10 mM Tris–HCl [pH 8.0], 150 mM NaCl, 1 mM EDTA, 0.5% NP-40, and proteinase inhibitor cocktail (Roche)) at 4 °C on rocking platform for 30 min. After centrifugation at 12,000g, the supernatant was incubated with specific antibodies against purified ALKBH2 and Protein A beads (Santa Cruz Biotechnology) for 16 h. Immunoprecipitated samples were loaded on 4 to 12% gradient SDS-PAGE gels, and gels were stained in Coomassie Brilliant Blue solution. Specific bands that were not found in the immunoglobulin G control lane were excised, trypsinized, and identified through the Mass Spectrometry and Proteomics Core at the City of Hope using a Thermo Fisher Scientific LTQ-FT MS that was coupled to an Eksigent NanoLC-2D HPLC system. Potential interactions from MS data were confirmed by immunoprecipitation and Western blotting with specific antibodies.

Generating KOs in HAP1 cells

Genes were targeted using a dual single-guide RNA CRISPR/Cas9 targeting vector to introduce deletions at adjacent targeting sites (40–80 bp separation) present in a single exon (Fig. 1A). MLH1ko, ALKBH2ko, ALKBH3ko, ALKBH2koALKBH3ko, MLH1koALKBH2ko, MLH1koALKBH3ko, and MLH1koALKBH2ko3ko HAP1 cells were derived using CRISPR/Cas9 gene targeting. Targeting vectors were designed to induce cleavage at exon 2 on ALKBH2, exon 2 of ALKBH3, and exon 12 of MLH1. Single-guide RNAs were designed using the online resource crispor.tefor.net (75), and cloned into the pX333 (plasmid 64073; Addgene) (76). Cells were transiently cotransfected with targeting vector and pmaxGFP vector (Lonza) using Xfect Transfection Reagent (Takara) according to manufacturer’s instructions. After transfection, individual cells were sorted into 96-well plates using flow cytometry (BD FACSAria II, BD Biosciences). Single cell colonies were cultured for up to 10 days following sorting. DNA was extracted from individual clones using Bradley lysis buffer (10 mM Tris–HCl [pH 7.5], 10 mM EDTA, 0.5% SDS, 10 mM NaCl, 1 mg/ml proteinase K, 0.4 mg/ml RNase) by incubating cells overnight at 37 °C. The following day, DNA was precipitated with glycogen, ammonium acetate (7.5 M), and 100% ethanol at −80 °C. Clones were selected for deletions following PCR using primers flanking the target sites on ALKBH2, ALKBH3, and MLH1. Clones identified with a positive PCR screen were further screened using Sanger sequencing to identify frame-shift mutations, and Western blot to verify loss of ALKBH2, ALKBH3, or MLH1 protein levels (Figs. 1, B and C and S4A).

Preparation of whole-cell extract

Total cell extracts were prepared using the rapid immune-filter paper assay lysis buffer system (Santa Cruz Biotechnology). Briefly, harvested cells were lysed using ice-cold rapid immune-filter paper assay lysis buffer containing protease inhibitors for 20 min at 4 °C and centrifuged at 14,000g for 15 min. Protein concentrations were determined using Bradford assay. Equal protein concentrations of protein samples were separated by denaturing SDS-PAGE and transferred onto a nitrocellulose membrane. Membranes were visualized using an Odyssey CLx Imaging System (Li-Cor).

Clonogenic and cell survival assay

For clonogenic assays, 100 cells were seeded in collagen coated 12-well plates. For cell survival assay, 15,000 cells were seeded in collagen coated 24-well plates. Cells were then exposed to serum-free DMEM containing increasing concentrations of either TMZ or MMS for 1 h at 37 °C. Drug treatment was then replaced with complete media, and cells were assessed at 7 or 3 days after treatment for clonogenic and cell survival, respectively. Briefly, cells were fixed with 10% neutral buffered formalin (Thermo Fisher Scientific) and stained with 0.01% (w/v) crystal violet. For clonogenic assays, individual colonies were counted, and relative survival was calculated by setting untreated controls as baseline. For the cell survival assay, crystal violet was solubilized with 10% v/v acetic acid, and the absorbance at 570 nm was determined with a Synergy HTX Multi-Mode Reader (BioTek), and absorbance of untreated controls were set as baseline. All clonogenic and cell survival experiments were carried out in triplicate.

Cell cycle analysis and determination of apoptosis

For BrdU labeling, cells were pulse-labeled with 10 μM BrdU (Sigma-Aldrich) for 30 min before harvesting and fixed in chilled 70% ethanol on ice for 1 h. The cells were processed as described (77). Cells were incubated in 2 N HCl/0.5% Triton X-100 for 30 min, neutralized with 0.1 M Borax for 2 min, and washed with PBS/1% bovine serum albumin. Samples were incubated in anti-BrdU (Novus) for 1 h, washed, treated with secondary antibody anti-rat AlexaFluor-488 (Thermo Fisher Scientific) for 30 min. Cells were washed and resuspended in PBS containing propidium iodide (20 μg/ml) and RNase A (10 μg/ml) for 30 min at room temperature. BrdU incorporation and cell cycle distributions were analyzed using BD Accuri C6 Flow Cytometer (Becton Dickinson) with >50,000 events per determination. Data were analyzed using FCS Express 7 (DeNovo Software, https://denovosoftware.com/).

Apoptotic frequency was determined using the FITC Annexin V Apoptosis Detection Kit II (Becton Dickinson) following manufacturer’s protocol and quantified by flow cytometry with C6 Accuri Flow Cytometer (Becton Dickinson) with 10,000 events per determination.

Mutant frequency assay

In total, 500,000 cells were treated with 50 μM TMZ for 1 h in serum-free DMEM. After treatment, cells were incubated for 5 days to allow for expression of ouabain-resistant mutations. Subsequently, 1 × 106 cells were incubated in complete media containing 1 μM ouabain for 10 days. Cells were fixed and stained, and the number of ouabain-resistant colonies were counted. In parallel, a cell suspension containing 100 cells was plated in six well plates, and the number of viable cells was counted to determine plating efficiency.

Immunofluorescence

Cells were plated in chamber slides at a density of 5.5 × 103 cells/cm2. After overnight cell adherence, the cells were treated with 100 μM TMZ for 1 h at 37 °C in IMDM media without FBS. After treatment, cells were washed with PBS and IMDM media +10% FBS added. Cells were incubated for 24 h. Following incubation, cells were fixed with 4% paraformaldehyde for 15 min at room temperature. Cells were then washed twice with PBS. Paraformaldehyde was quenched with 0.1 M glycine in PBS for 10 min and then washed twice with PBS. Cells were permeabilized with 0.5% Triton for 15 min and then washed twice with PBS. Blocking solution (2% bovine serum albumin in PBS) was added for 30 min at room temperature with rocking. Primary antibodies (γ-H2AX: Novus NB100-78356, 1:500 and 53BP1: Abcam ab36823, 1:1000) were added overnight in a 37 °C incubator. Cells were then washed three times with PBS. Secondary antibodies (Alexa Fluor 488 donkey anti-mouse: Invitrogen A11029, 1:250 and Alexa Fluor 594 goat anti-rabbit: Invitrogen A11037, 1:250) were added for 1 h in a 37 °C incubator. Cells were then washed three times with PBS. Coverslips were mounted using VECTASHIELD (Vector laboratories) and dried for 2 hours in the dark. Images were obtained on LSM 880 AxioObserver with a Plan-Apochromat 63×/1.4 oil DIC M27 objective. Green and red channels were overlaid and colocalization was determined by the presence of yellow dots with manual counting of the number of colocalization events within a cell.

DNA fiber assays

HAP1 cells were plated 24 h prior to treatment. Cells were labeled with 50 μM IdU (Sigma-Aldrich) for 30 min in complete media. IdU was then replaced with media containing 250 μM CldU (Sigma-Aldrich) with either DMSO or 500 μM TMZ. CldU labeling and treatment was terminated after 30 min. Cells were then washed with ice-cold PBS-DMEM, harvested via trypsinization, and counted. A total of 6.0 × 105 cells were collected and used for DNA fiber analysis. DNA was extracted and spread onto a silanized coverslip in accordance with the FiberPrep: DNA Extraction Kit protocol (Genomic Vision). Coverslips were then air dried, and DNA fibers were fixed onto coverslips with 0.5 M NaOH, 1 M NaCl solution. Fibers were stained with mouse anti BrdU (BD) and rat anti Bardu (Novus) for 2 h. Fibers were then stained with secondary antibodies, rabbit anti-mouse AlexaFluor-594 and chicken anti-rat AlexaFluor-488 for 1 h, followed by tertiary antibodies, goat anti-rabbit AlexaFluor-555 and goat anti-chicken- AlexaFluor-488, for 1 h. DNA fibers were observed under a Zeiss Observer 2 light microscope. The lengths of CldU and IdU tracks were measured using the Zen 2.6 (Blue Edition, https://www.zeiss.com/microscopy/us/products/software/zeiss-zen.html) software. At least 100 forks were analyzed for every condition.

Mass spectrometry analysis

Proteins were separated on a 10% SDS-PAGE gel, gel bands excised and reduced with DTT, alkylated with iodoacetamide, and digested using trypsin. Analysis was performed on an LTQ-FT MS equipped with an Eksigent NanoLC 2D. Samples were loaded onto an LC Packing 300 μm × 5 mm trapping column at 10 μl/min for 5 minutes. The sample was eluted onto a self-packed 75 μm × 10 cm column packed with Varian Extend 3 μm C18. The gradient was run using mobile phase A: H2O + 0.1% formic acid and mobile phase B: acetonitrile with 0.1% formic acid at a rate of 200 nl/min with the following gradient: 0 to 3 min, 5% B; 3 to 6 min, 11% B; 6 to 48 min, 35% B; 48 to 52 min, 55% B; 52 to 54 min, 95% B; 54 to 57 min, 95% B; 57 to 60 min, 5% B; and 60 to 65 min, 5% B. MS was performed in the FT cell scanning from 300 to 2000 at 100,000 nominal resolution. Data-dependent MS/MS was performed on the top five ions from the precursor scan. Dynamic exclusion after 1 MS/MS with a 40 s duration was used. Data were searched using TurboSequest v.27 (http://tools.proteomecenter.org) against the NCBI NR database with trypsin specificity, no more than two missed cleavages, quantitative carbamidomethylation on cysteine, and differential oxidation on methionine. Search results were processed using Qscore.

Data availability

Data will be made available upon request.

Conflict of interest

The authors declare that they have no conflicts of interest with the contents of this article.

Supporting information

This article contains supporting information.

Acknowledgments

The authors acknowledge the discussions with Drs. Jeremy Stark, Yilun Liu, John Termini, and WenYong Chen.

Author contributions

R. G., A. Y. S. C., S. W. T. L., S. I., D.-H. L., K. S., A. B., S. C., T. R. O. C., and S. C. S. formal analysis; R. G., A. Y. S. C., S. W. T. L., S. I., D.-H. L., K. S., A. B., T. R. O. C., and S. C. S. data curation; R. G., A. Y. S. C., T. R. O. C., and S. C. S. methodology; R. G., S. C.., T. R. O. C., and S. C. S. resources; R. G., T. R. O. C., and S. C. S. investigation; R. G., T. R. O. C. and S. C. S. conceptualization; R. G., T. R. O. C. and S. C. S. writing–original draft; R. G., T. R. O. C. and S. C. S. writing–review and editing; S. C., T. R. O. C., and S. C. S. project administration; S. C., T. R. O. C. and S. C. S. supervision; T. R. O. C. and S. C. S. validation; T. R. O. C. and S. C. S. visualization; T. R. O. C. funding acquisition.

Funding and additional information

This work was supported by the California Tobacco-Related Disease Research Program [(No. 28IR-0050, T. R. O. C.)], and the Irell & Manella Graduate School of Biological Sciences: Payson and Chu Fellowship [R. G.]. Research reported in this publication included work performed in the integrative genomics, analytical cytometry, and light microscopy core supported by the National Cancer Institutes of Health under grant number P30CA033572. The content is solely the responsibility of the authors and does not necessarily represent the official of the National Institutes of Health.

Notes

Reviewed by members of the JBC Editorial Board. Edited by Patrick Sung

Footnotes

Present addresses for: Dong-Hyun Lee, Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju 61186 South Korea; Shunsuke Itoh, School of Medicine, Yamaguchi University, Ube City, Yamaguchi -Prefecture 755 to 8505, Japan.

Supporting information

Supplemental Figures:

References

1. Warwick G.P. The mechanism of action of alkylating agents. Cancer Res. 1963;23:1315–1333. [Abstract] [Google Scholar]
2. Fu D., Calvo J.A., Samson L.D. Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat. Rev. Cancer. 2012;12:104–120. [Europe PMC free article] [Abstract] [Google Scholar]
3. Scharer O.D., Jiricny J. Recent progress in the biology, chemistry and structural biology of DNA glycosylases. Bioessays. 2001;23:270–281. [Abstract] [Google Scholar]
4. Seeberg E., Eide L., Bjoras M. The base excision repair pathway. Trends Biochem. Sci. 1995;20:391–397. [Abstract] [Google Scholar]
5. Falnes P.O., Johansen R.F., Seeberg E. AlkB-mediated oxidative demethylation reverses DNA damage in Escherichia coli. Nature. 2002;419:178–182. [Abstract] [Google Scholar]
6. Trewick S.C., Henshaw T.F., Hausinger R.P., Lindahl T., Sedgwick B. Oxidative demethylation by Escherichia coli AlkB directly reverts DNA base damage. Nature. 2002;419:174–178. [Abstract] [Google Scholar]
7. Delaney J.C., Essigmann J.M. Mutagenesis, genotoxicity, and repair of 1-methyladenine, 3-alkylcytosines, 1-methylguanine, and 3-methylthymine in alkB Escherichia coli. Proc. Natl. Acad. Sci. U. S. A. 2004;101:14051–14056. [Europe PMC free article] [Abstract] [Google Scholar]
8. Dinglay S., Trewick S.C., Lindahl T., Sedgwick B. Defective processing of methylated single-stranded DNA by E. coli AlkB mutants. Genes Dev. 2000;14:2097–2105. [Europe PMC free article] [Abstract] [Google Scholar]
9. Duncan T., Trewick S.C., Koivisto P., Bates P.A., Lindahl T., Sedgwick B. Reversal of DNA alkylation damage by two human dioxygenases. Proc. Natl. Acad. Sci. U. S. A. 2002;99:16660–16665. [Europe PMC free article] [Abstract] [Google Scholar]
10. Aas P.A., Otterlei M., Falnes P.O., Vagbo C.B., Skorpen F., Akbari M., et al. Human and bacterial oxidative demethylases repair alkylation damage in both RNA and DNA. Nature. 2003;421:859–863. [Abstract] [Google Scholar]
11. Fu D., Samson L.D., Hübscher U., van Loon B. The interaction between ALKBH2 DNA repair enzyme and PCNA is direct, mediated by the hydrophobic pocket of PCNA and perturbed in naturally-occurring ALKBH2 variants. DNA Repair. 2015;35:13–18. [Europe PMC free article] [Abstract] [Google Scholar]
12. Tsujikawa K., Koike K., Kitae K., Shinkawa A., Arima H., Suzuki T., et al. Expression and sub-cellular localization of human ABH family molecules. J. Cell. Mol. Med. 2007;11:1105–1116. [Europe PMC free article] [Abstract] [Google Scholar]
13. Falnes P.O., Bjoras M., Aas P.A., Sundheim O., Seeberg E. Substrate specificities of bacterial and human AlkB proteins. Nucleic Acids Res. 2004;32:3456–3461. [Europe PMC free article] [Abstract] [Google Scholar]
14. Dango S., Mosammaparast N., Sowa M.E., Xiong L.J., Wu F., Park K., et al. DNA unwinding by ASCC3 helicase is coupled to ALKBH3-dependent DNA alkylation repair and cancer cell proliferation. Mol. Cell. 2011;44:373–384. [Europe PMC free article] [Abstract] [Google Scholar]
15. Furrer A., van Loon B. Handling the 3-methylcytosine lesion by six human DNA polymerases members of the B-, X- and Y-families. Nucleic Acids Res. 2014;42:553–566. [Europe PMC free article] [Abstract] [Google Scholar]
16. Conde J., Yoon J.H., Roy Choudhury J., Prakash L., Prakash S. Genetic control of replication through N1-methyladenine in human cells. J. Biol. Chem. 2015;290:29794–29800. [Europe PMC free article] [Abstract] [Google Scholar]
17. Nay S.L., Lee D.H., Bates S.E., O'Connor T.R. Alkbh2 protects against lethality and mutation in primary mouse embryonic fibroblasts. DNA Repair. 2012;11:502–510. [Europe PMC free article] [Abstract] [Google Scholar]
18. Ringvoll J., Nordstrand L.M., Vagbo C.B., Talstad V., Reite K., Aas P.A., et al. Repair deficient mice reveal mABH2 as the primary oxidative demethylase for repairing 1meA and 3meC lesions in DNA. EMBO J. 2006;25:2189–2198. [Europe PMC free article] [Abstract] [Google Scholar]
19. Huang Y., Li G.M. DNA mismatch repair preferentially safeguards actively transcribed genes. DNA Repair. 2018;71:82–86. [Europe PMC free article] [Abstract] [Google Scholar]
20. Klapacz J., Pottenger L.H., Engelward B.P., Heinen C.D., Johnson G.E., Clewell R.A., et al. Contributions of DNA repair and damage response pathways to the non-linear genotoxic responses of alkylating agents. Mutat. Res. Rev. Mutat. Res. 2016;767:77–91. [Europe PMC free article] [Abstract] [Google Scholar]
21. Li G.M. The role of mismatch repair in DNA damage-induced apoptosis. Oncol. Res. 1999;11:393–400. [Abstract] [Google Scholar]
22. Duckett D.R., Drummond J.T., Murchie A.I., Reardon J.T., Sancar A., Lilley D.M., et al. Human MutSalpha recognizes damaged DNA base pairs containing O6-methylguanine, O4-methylthymine, or the cisplatin-d(GpG) adduct. Proc. Natl. Acad. Sci. U. S. A. 1996;93:6443–6447. [Europe PMC free article] [Abstract] [Google Scholar]
23. Wu J., Gu L., Wang H., Geacintov N.E., Li G.M. Mismatch repair processing of carcinogen-DNA adducts triggers apoptosis. Mol. Cell Biol. 1999;19:8292–8301. [Europe PMC free article] [Abstract] [Google Scholar]
24. Simonelli V., Leuzzi G., Basile G., D'Errico M., Fortini P., Franchitto A., et al. Crosstalk between mismatch repair and base excision repair in human gastric cancer. Oncotarget. 2017;8:84827–84840. [Europe PMC free article] [Abstract] [Google Scholar]
25. Mu D., Tursun M., Duckett D.R., Drummond J.T., Modrich P., Sancar A. Recognition and repair of compound DNA lesions (base damage and mismatch) by human mismatch repair and excision repair systems. Mol. Cell Biol. 1997;17:760–769. [Europe PMC free article] [Abstract] [Google Scholar]
26. York S.J., Modrich P. Mismatch repair-dependent iterative excision at irreparable O6-methylguanine lesions in human nuclear extracts. J. Biol. Chem. 2006;281:22674–22683. [Europe PMC free article] [Abstract] [Google Scholar]
27. Stojic L., Brun R., Jiricny J. Mismatch repair and DNA damage signalling. DNA Repair. 2004;3:1091–1101. [Abstract] [Google Scholar]
28. Colussi C., Parlanti E., Degan P., Aquilina G., Barnes D., Macpherson P., et al. The mammalian mismatch repair pathway removes DNA 8-oxodGMP incorporated from the oxidized dNTP pool. Curr. Biol. 2002;12:912–918. [Abstract] [Google Scholar]
29. Russo M.T., De Luca G., Casorelli I., Degan P., Molatore S., Barone F., et al. Role of MUTYH and MSH2 in the control of oxidative DNA damage, genetic instability, and tumorigenesis. Cancer Res. 2009;69:4372–4379. [Abstract] [Google Scholar]
30. Mazurek A., Berardini M., Fishel R. Activation of human MutS homologs by 8-oxo-guanine DNA damage. J. Biol. Chem. 2002;277:8260–8266. [Abstract] [Google Scholar]
31. Edelbrock M.A., Kaliyaperumal S., Williams K.J. Structural, molecular and cellular functions of MSH2 and MSH6 during DNA mismatch repair, damage signaling and other noncanonical activities. Mutat. Res. 2013;743-744:53–66. [Europe PMC free article] [Abstract] [Google Scholar]
32. Stojic L., Mojas N., Cejka P., Di Pietro M., Ferrari S., Marra G., et al. Mismatch repair-dependent G2 checkpoint induced by low doses of SN1 type methylating agents requires the ATR kinase. Genes Dev. 2004;18:1331–1344. [Europe PMC free article] [Abstract] [Google Scholar]
33. Takagi Y., Takahashi M., Sanada M., Ito R., Yamaizumi M., Sekiguchi M. Roles of MGMT and MLH1 proteins in alkylation-induced apoptosis and mutagenesis. DNA Repair. 2003;2:1135–1146. [Abstract] [Google Scholar]
34. Branch P., Aquilina G., Bignami M., Karran P. Defective mismatch binding and a mutator phenotype in cells tolerant to DNA damage. Nature. 1993;362:652–654. [Abstract] [Google Scholar]
35. Kat A., Thilly W.G., Fang W.H., Longley M.J., Li G.M., Modrich P. An alkylation-tolerant, mutator human cell line is deficient in strand-specific mismatch repair. Proc. Natl. Acad. Sci. U. S. A. 1993;90:6424–6428. [Europe PMC free article] [Abstract] [Google Scholar]
36. Kawate H., Sakumi K., Tsuzuki T., Nakatsuru Y., Ishikawa T., Takahashi S., et al. Separation of killing and tumorigenic effects of an alkylating agent in mice defective in two of the DNA repair genes. Proc. Natl. Acad. Sci. U. S. A. 1998;95:5116–5120. [Europe PMC free article] [Abstract] [Google Scholar]
37. Ochs K., Kaina B. Apoptosis induced by DNA damage O6-methylguanine is Bcl-2 and caspase-9/3 regulated and Fas/caspase-8 independent. Cancer Res. 2000;60:5815–5824. [Abstract] [Google Scholar]
38. Hunter C., Smith R., Cahill D.P., Stephens P., Stevens C., Teague J., et al. A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy. Cancer Res. 2006;66:3987–3991. [Europe PMC free article] [Abstract] [Google Scholar]
39. Yip S., Miao J., Cahill D.P., Iafrate A.J., Aldape K., Nutt C.L., et al. MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin. Cancer Res. 2009;15:4622–4629. [Europe PMC free article] [Abstract] [Google Scholar]
40. Daniel P., Sabri S., Chaddad A., Meehan B., Jean-Claude B., Rak J., et al. Temozolomide induced hypermutation in glioma: evolutionary mechanisms and therapeutic opportunities. Front. Oncol. 2019;9:41. [Europe PMC free article] [Abstract] [Google Scholar]
41. McCord M., Steffens A., Javier R., Kam K.L., McCortney K., Horbinski C. The efficacy of DNA mismatch repair enzyme immunohistochemistry as a screening test for hypermutated gliomas. Acta Neuropathol. Commun. 2020;8:15. [Europe PMC free article] [Abstract] [Google Scholar]
42. Wittmeyer J., Formosa T. The Saccharomyces cerevisiae DNA polymerase alpha catalytic subunit interacts with Cdc68/Spt16 and with Pob3, a protein similar to an HMG1-like protein. Mol. Cell Biol. 1997;17:4178–4190. [Europe PMC free article] [Abstract] [Google Scholar]
43. Keller D.M., Lu H. p53 serine 392 phosphorylation increases after UV through induction of the assembly of the CK2.hSPT16.SSRP1 complex. J. Biol. Chem. 2002;277:50206–50213. [Abstract] [Google Scholar]
44. Heo K., Kim H., Choi S.H., Choi J., Kim K., Gu J., et al. FACT-mediated exchange of histone variant H2AX regulated by phosphorylation of H2AX and ADP-ribosylation of Spt16. Mol. Cell. 2008;30:86–97. [Abstract] [Google Scholar]
45. Niture S.K., Doneanu C.E., Velu C.S., Bailey N.I., Srivenugopal K.S. Proteomic analysis of human O6-methylguanine-DNA methyltransferase by affinity chromatography and tandem mass spectrometry. Biochem. Biophys. Res. Commun. 2005;337:1176–1184. [Abstract] [Google Scholar]
46. Buermeyer A.B., Wilson-Van Patten C., Baker S.M., Liskay R.M. The human MLH1 cDNA complements DNA mismatch repair defects in Mlh1-deficient mouse embryonic fibroblasts. Cancer Res. 1999;59:538–541. [Abstract] [Google Scholar]
47. Drabløs F., Feyzi E., Aas P.A., Vaagbø C.B., Kavli B., Bratlie M.S., et al. Alkylation damage in DNA and RNA--repair mechanisms and medical significance. DNA Repair. 2004;3:1389–1407. [Abstract] [Google Scholar]
48. Begley T.J., Samson L.D. AlkB mystery solved: oxidative demethylation of N1-methyladenine and N3-methylcytosine adducts by a direct reversal mechanism. Trends Biochem. Sci. 2003;28:2–5. [Abstract] [Google Scholar]
49. Li G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008;18:85–98. [Abstract] [Google Scholar]
50. Liu A., Yoshioka K., Salerno V., Hsieh P. The mismatch repair-mediated cell cycle checkpoint response to fluorodeoxyuridine. J. Cell Biochem. 2008;105:245–254. [Europe PMC free article] [Abstract] [Google Scholar]
51. Boiteux S., Laval J. Mutagenesis by alkylating agents: coding properties for DNA polymerase of poly (dC) template containing 3-methylcytosine. Biochimie. 1982;64:637–641. [Abstract] [Google Scholar]
52. Larson K., Sahm J., Shenkar R., Strauss B. Methylation-induced blocks to in vitro DNA replication. Mutat. Res. 1985;150:77–84. [Abstract] [Google Scholar]
53. Wang P., Wu J., Ma S., Zhang L., Yao J., Hoadley K.A., et al. Oncometabolite D-2-hydroxyglutarate inhibits ALKBH DNA repair enzymes and sensitizes IDH mutant cells to alkylating agents. Cell Rep. 2015;13:2353–2361. [Europe PMC free article] [Abstract] [Google Scholar]
54. Lobrich M., Shibata A., Beucher A., Fisher A., Ensminger M., Goodarzi A.A., et al. gammaH2AX foci analysis for monitoring DNA double-strand break repair: strengths, limitations and optimization. Cell Cycle. 2010;9:662–669. [Abstract] [Google Scholar]
55. Tewari M., Quan L.T., O'Rourke K., Desnoyers S., Zeng Z., Beidler D.R., et al. Yama/CPP32 beta, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase. Cell. 1995;81:801–809. [Abstract] [Google Scholar]
56. Lin J.K., Kuo M.L. Induction of ouabain-resistance mutation and cycle-dependent transformation of C3H/10T1/2 cells by N-nitroso-2-acetylaminofluorene. Mutat. Res. 1990;230:35–43. [Abstract] [Google Scholar]
57. Meikrantz W., Bergom M.A., Memisoglu A., Samson L. O6-alkylguanine DNA lesions trigger apoptosis. Carcinogenesis. 1998;19:369–372. [Abstract] [Google Scholar]
58. Rasmussen L.J., Samson L. The Escherichia coli MutS DNA mismatch binding protein specifically binds O(6)-methylguanine DNA lesions. Carcinogenesis. 1996;17:2085–2088. [Abstract] [Google Scholar]
59. Dosch J., Christmann M., Kaina B. Mismatch G-T binding activity and MSH2 expression is quantitatively related to sensitivity of cells to methylating agents. Carcinogenesis. 1998;19:567–573. [Abstract] [Google Scholar]
60. Hickman M.J., Samson L.D. Role of DNA mismatch repair and p53 in signaling induction of apoptosis by alkylating agents. Proc. Natl. Acad. Sci. U. S. A. 1999;96:10764–10769. [Europe PMC free article] [Abstract] [Google Scholar]
61. Hampson R., Humbert O., Macpherson P., Aquilina G., Karran P. Mismatch repair defects and O6-methylguanine-DNA methyltransferase expression in acquired resistance to methylating agents in human cells. J. Biol. Chem. 1997;272:28596–28606. [Abstract] [Google Scholar]
62. D'Atri S., Tentori L., Lacal P.M., Graziani G., Pagani E., Benincasa E., et al. Involvement of the mismatch repair system in temozolomide-induced apoptosis. Mol. Pharmacol. 1998;54:334–341. [Abstract] [Google Scholar]
63. Bearzatto A., Szadkowski M., Macpherson P., Jiricny J., Karran P. Epigenetic regulation of the MGMT and hMSH6 DNA repair genes in cells resistant to methylating agents. Cancer Res. 2000;60:3262–3270. [Abstract] [Google Scholar]
64. Merrick C.J., Jackson D., Diffley J.F. Visualization of altered replication dynamics after DNA damage in human cells. J. Biol. Chem. 2004;279:20067–20075. [Abstract] [Google Scholar]
65. Groth P., Ausländer S., Majumder M.M., Schultz N., Johansson F., Petermann E., et al. Methylated DNA causes a physical block to replication forks independently of damage signalling, O6-methylguanine or DNA single-strand breaks and results in DNA damage. J. Mol. Biol. 2010;402:70–82. [Abstract] [Google Scholar]
66. Beranek D.T. Distribution of methyl and ethyl adducts following alkylation with monofunctional alkylating agents. Mutat. Res. 1990;231:11–30. [Abstract] [Google Scholar]
67. Yoshioka K., Yoshioka Y., Hsieh P. ATR kinase activation mediated by MutSalpha and MutLalpha in response to cytotoxic O6-methylguanine adducts. Mol. Cell. 2006;22:501–510. [Europe PMC free article] [Abstract] [Google Scholar]
68. Sansom O.J., Toft N.J., Winton D.J., Clarke A.R. Msh-2 suppresses in vivo mutation in a gene dose and lesion dependent manner. Oncogene. 2001;20:3580–3584. [Abstract] [Google Scholar]
69. Bugni J.M., Meira L.B., Samson L.D. Alkylation-induced colon tumorigenesis in mice deficient in the Mgmt and Msh6 proteins. Oncogene. 2009;28:734–741. [Europe PMC free article] [Abstract] [Google Scholar]
70. Kawate H., Itoh R., Sakumi K., Nakabeppu Y., Tsuzuki T., Ide F., et al. A defect in a single allele of the Mlh1 gene causes dissociation of the killing and tumorigenic actions of an alkylating carcinogen in methyltransferase-deficient mice. Carcinogenesis. 2000;21:301–305. [Abstract] [Google Scholar]
71. Hewish M., Lord C.J., Martin S.A., Cunningham D., Ashworth A. Mismatch repair deficient colorectal cancer in the era of personalized treatment. Nat. Rev. Clin. Oncol. 2010;7:197–208. [Abstract] [Google Scholar]
72. Kaina B., Fritz G., Mitra S., Coquerelle T. Transfection and expression of human O6-methylguanine-DNA methyltransferase (MGMT) cDNA in Chinese hamster cells: the role of MGMT in protection against the genotoxic effects of alkylating agents. Carcinogenesis. 1991;12:1857–1867. [Abstract] [Google Scholar]
73. Lips J., Kaina B. Repair of O(6)-methylguanine is not affected by thymine base pairing and the presence of MMR proteins. Mutat. Res. 2001;487:59–66. [Abstract] [Google Scholar]
74. Essletzbichler P., Konopka T., Santoro F., Chen D., Gapp B.V., Kralovics R., et al. Megabase-scale deletion using CRISPR/Cas9 to generate a fully haploid human cell line. Genome Res. 2014;24:2059–2065. [Europe PMC free article] [Abstract] [Google Scholar]
75. Haeussler M., Schonig K., Eckert H., Eschstruth A., Mianne J., Renaud J.B., et al. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biol. 2016;17:148. [Europe PMC free article] [Abstract] [Google Scholar]
76. Maddalo D., Manchado E., Concepcion C.P., Bonetti C., Vidigal J.A., Han Y.C., et al. In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature. 2014;516:423–427. [Europe PMC free article] [Abstract] [Google Scholar]
77. Cliby W.A., Lewis K.A., Lilly K.K., Kaufmann S.H. S phase and G2 arrests induced by topoisomerase I poisons are dependent on ATR kinase function. J. Biol. Chem. 2002;277:1599–1606. [Abstract] [Google Scholar]

Articles from The Journal of Biological Chemistry are provided here courtesy of American Society for Biochemistry and Molecular Biology

Citations & impact 


This article has not been cited yet.

Impact metrics

Alternative metrics

Altmetric item for https://www.altmetric.com/details/165059876
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/165059876

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

NCI NIH HHS (1)