Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter June 30, 2021

Structural insights into the repair mechanism of AGT for methyl-induced DNA damage

  • Rajendra P. Koirala , Rudramani Pokhrel , Prabin Baral , Purushottam B. Tiwari ORCID logo EMAIL logo , Prem P. Chapagain ORCID logo EMAIL logo and Narayan P. Adhikari ORCID logo EMAIL logo
From the journal Biological Chemistry

Abstract

Methylation induced DNA base-pairing damage is one of the major causes of cancer. O6-alkylguanine-DNA alkyltransferase (AGT) is considered a demethylation agent of the methylated DNA. Structural investigations with thermodynamic properties of the AGT-DNA complex are still lacking. In this report, we modeled two catalytic states of AGT-DNA interactions and an AGT-DNA covalent complex and explored structural features using molecular dynamics (MD) simulations. We utilized the umbrella sampling method to investigate the changes in the free energy of the interactions in two different AGT-DNA catalytic states, one with methylated GUA in DNA and the other with methylated CYS145 in AGT. These non-covalent complexes represent the pre- and post-repair complexes. Therefore, our study encompasses the process of recognition, complex formation, and separation of the AGT and the damaged (methylated) DNA base. We believe that the use of parameters for the amino acid and nucleotide modifications and for the protein-DNA covalent bond will allow investigations of the DNA repair mechanism as well as the exploration of cancer therapeutics targeting the AGT-DNA complexes at various functional states as well as explorations via stabilization of the complex.


Corresponding authors: Purushottam B. Tiwari, Department of Oncology, Georgetown University, Washington, D.C., USA, E-mail: ; Prem P. Chapagain, Department of Physics, Florida International University, Miami, FL, USA; and Biomolecular Sciences Institute, Florida International University, Miami, FL, USA, E-mail: ; and Narayan P. Adhikari, Central Department of Physics, Tribhuvan University, Kathmandu, Nepal, E-mail:

Award Identifier / Grant number: CRG-73/74-S&T-01

Acknowledgments

We sincerely thank Professor Alexander D. MacKerell Jr. at the Computer-Aided Drug Design Center, University of Maryland, USA, for his valuable input and help with the force-field parameters. R.P.K. acknowledges the partial financial support from the Nepal Academy of Science and Technology (NAST). N.P.A. acknowledges the UGC Award No. CRG-73/74-S&T-01 and Associate membership of The Abdus Salam International Center for Theoretical Physics, Trieste, Italy.

  1. Author contributions: P.B.T., P.P.C., and N.P.A. guided the project. R.P.K, R.P., P.B., and P.B.T performed MD simulations. R.P.K., R.P., P.B., P.B.T., P.P.C., and N.P.A. analyzed the data. R.P.K., P.B.T., and P.P.C. wrote the manuscript. R.P., P.B., and N.P.A. contributed to the manuscript editing.

  2. Research funding: This work was supported by University Grants Commission- Nepal (CRG-73/74-S&T-01).

  3. Conflict of interest statement: The authors declare no conflict of interest.

References

Ali, R.B., Teo, A.K., Oh, H.K., Chuang, L.S., Ayi, T.C., and Li, B.F. (1998). Implication of localization of human DNA repair enzyme O6-methylguanine-DNA methyltransferase at active transcription sites in transcription-repair coupling of the mutagenic O6-methylguanine lesion. Mol. Cell. Biol. 18: 1660–1669, https://doi.org/10.1128/mcb.18.3.1660.Search in Google Scholar

Banavali, N.K. and MacKerell, A.D.Jr. (2002). Free energy and structural pathways of base flipping in a DNA GCGC containing sequence. J. Mol. Biol. 319: 141–160, https://doi.org/10.1016/s0022-2836(02)00194-8.Search in Google Scholar

Becker, K., Dosch, J., Gregel, C.M., Martin, B.A., and Kaina, B. (1996). Targeted expression of human O(6)-methylguanine-DNA methyltransferase (MGMT) in transgenic mice protects against tumor initiation in two-stage skin carcinogenesis. Canc. Res. 56: 3244–3249.Search in Google Scholar

Crone, T.M., Goodtzova, K., and Pegg, A.E. (1996). Amino acid residues affecting the activity and stability of human O6-alkylguanine-DNA alkyltransferase. Mutat. Res. 363: 15–25, https://doi.org/10.1016/0921-8777(95)00058-5.Search in Google Scholar

Daniels, D.S., Mol, C.D., Arvai, A.S., Kanugula, S., Pegg, A.E., and Tainer, J.A. (2000). Active and alkylated human AGT structures: a novel zinc site, inhibitor and extrahelical base binding. EMBO J. 19: 1719–1730, https://doi.org/10.1093/emboj/19.7.1719.Search in Google Scholar

Daniels, D.S., Woo, T.T., Luu, K.X., Noll, D.M., Clarke, N.D., Pegg, A.E., and Tainer, J.A. (2004). DNA binding and nucleotide flipping by the human DNA repair protein AGT. Nat. Struct. Mol. Biol. 11: 714–720, https://doi.org/10.1038/nsmb791.Search in Google Scholar

De Bont, R. and van Larebeke, N. (2004). Endogenous DNA damage in humans: a review of quantitative data. Mutagenesis 19: 169–185, https://doi.org/10.1093/mutage/geh025.Search in Google Scholar

Duguid, E.M., Rice, P.A., and He, C. (2005). The structure of the human AGT protein bound to DNA and its implications for damage detection. J. Mol. Biol. 350: 657–666, https://doi.org/10.1016/j.jmb.2005.05.028.Search in Google Scholar

Dumenco, L.L., Allay, E., Norton, K., and Gerson, S.L. (1993). The prevention of thymic lymphomas in transgenic mice by human O6-alkylguanine-DNA alkyltransferase. Science 259: 219–222, https://doi.org/10.1126/science.8421782.Search in Google Scholar

Fang, Q., Noronha, A.M., Murphy, S.P., Wilds, C.J., Tubbs, J.L., Tainer, J.A., Chowdhury, G., Guengerich, F.P., and Pegg, A.E. (2008). Repair of O6-G-alkyl-O6-G interstrand cross-links by human O6-alkylguanine-DNA alkyltransferase. Biochemistry 47: 10892–10903, https://doi.org/10.1021/bi8008664.Search in Google Scholar

Fernández, I., Uggerud, E., and Frenking, G. (2007). Stable pentacoordinate carbocations: structure and bonding. Chemistry 13: 8620–8626, https://doi.org/10.1002/chem.200700744.Search in Google Scholar

Fu, D., Calvo, J.A., and Samson, L.D. (2012). Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat. Rev. Canc. 12: 104–120, https://doi.org/10.1038/nrc3185.Search in Google Scholar

Fu, Y., Bernasconi, L., and Lu, P. (2021). Ab initio molecular dynamics simulations of the SN1/SN2 mechanistic continuum in glycosylation reactions. J. Am. Chem. Soc. 143: 1577–1589, https://doi.org/10.1021/jacs.0c12096.Search in Google Scholar

Gerson, S.L. (2002). Clinical relevance of MGMT in the treatment of cancer. J. Clin. Oncol. 20: 2388–2399, https://doi.org/10.1200/jco.2002.06.110.Search in Google Scholar

Harris, L.C., Potter, P.M., and Margison, G.P. (1992). Site directed mutagenesis of two cysteine residues in the E. coli OGT O6-alkylguanine DNA alkyltransferase protein. Biochem. Biophys. Res. Commun. 187: 425–431, https://doi.org/10.1016/s0006-291x(05)81510-4.Search in Google Scholar

Hu, J., Ma, A., and Dinner, A.R. (2008). A two-step nucleotide-flipping mechanism enables kinetic discrimination of DNA lesions by AGT. Proc. Natl. Acad. Sci. U. S. A. 105: 4615–4620, https://doi.org/10.1073/pnas.0708058105.Search in Google Scholar

Huang, J., Rauscher, S., Nawrocki, G., Ran, T., Feig, M., de Groot, B.L., Grubmüller, H., and MacKerell, A.D. (2017). CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14: 71–73, https://doi.org/10.1038/nmeth.4067.Search in Google Scholar

Humphrey, W., Dalke, A., and Schulten, K. (1996). VMD: visual molecular dynamics. J. Mol. Graph. 14: 27–28, https://doi.org/10.1016/0263-7855(96)00018-5.Search in Google Scholar

Jackson, S.P. and Bartek, J. (2009). The DNA-damage response in human biology and disease. Nature 461: 1071–1078, https://doi.org/10.1038/nature08467.Search in Google Scholar

Kaina, B., Christmann, M., Naumann, S., and Roos, W.P. (2007). MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair 6: 1079–1099, https://doi.org/10.1016/j.dnarep.2007.03.008.Search in Google Scholar

Kelley, M.R. and Fishel, M.L. (2008). DNA repair proteins as molecular targets for cancer therapeutics. Anti Canc. Agents Med. Chem. 8: 417–425, https://doi.org/10.2174/187152008784220294.Search in Google Scholar

Kumar, S., Rosenberg, J.M., Bouzida, D., Swendsen, R.H., and Kollman, P.A. (1992). The weighted histogram analysis method for free‐energy calculations on biomolecules. I. The method. J. Comput. Chem. 13: 1011–1021, https://doi.org/10.1002/jcc.540130812.Search in Google Scholar

Kyrtopoulos, S.A., Anderson, L.M., Chhabra, S.K., Souliotis, V.L., Pletsa, V., Valavanis, C., and Georgiadis, P. (1997). DNA adducts and the mechanism of carcinogenesis and cytotoxicity of methylating agents of environmental and clinical significance. Canc. Detect. Prev. 21: 391–405.Search in Google Scholar

Lee, J., Cheng, X., Swails, J.M., Yeom, M.S., Eastman, P.K., Lemkul, J.A., Wei, S., Buckner, J., Jeong, J.C., Qi, Y., et al.. (2016). CHARMM-GUI input generator for NAMD, GROMACS, AMBER, OpenMM, and CHARMM/OpenMM simulations using the CHARMM36 additive force field. J. Chem. Theor. Comput. 12: 405–413, https://doi.org/10.1021/acs.jctc.5b00935.Search in Google Scholar

Lindahl, T. and Barnes, D.E. (2000). Repair of endogenous DNA damage. Cold Spring Harbor Symp. Quant. Biol. 65: 127–133, https://doi.org/10.1101/sqb.2000.65.127.Search in Google Scholar

Liu, L., Qin, X., and Gerson, S.L. (1999). Reduced lung tumorigenesis in human methylguanine DNA--methyltransferase transgenic mice achieved by expression of transgene within the target cell. Carcinogenesis 20: 279–284, https://doi.org/10.1093/carcin/20.2.279.Search in Google Scholar

Luzhkov, V.B. (2017). Molecular modelling and free-energy calculations of protein—ligand binding. Russ. Chem. Rev. 86: 211, https://doi.org/10.1070/rcr4610.Search in Google Scholar

Magnen, M., Elsasser, B.M., Zbodakova, O., Kasparek, P., Gueugnon, F., Petit-Courty, A., Sedlacek, R., Goettig, P., and Courty, Y. (2018). Kallikrein-related peptidase 5 and seasonal influenza viruses, limitations of the experimental models for activating proteases. Biol. Chem. 399: 1053–1064, https://doi.org/10.1515/hsz-2017-0340.Search in Google Scholar

Maser, R.S. and DePinho, R.A. (2002). Connecting chromosomes, crisis, and cancer. Science 297: 565–569, https://doi.org/10.1126/science.297.5581.565.Search in Google Scholar

Mattossovich, R., Merlo, R., Miggiano, R., Valenti, A., and Perugino, G. (2020). O6-alkylguanine-DNA alkyltransferases in microbes living on the edge: from stability to applicability. Int. J. Mol. Sci. 21: 2878, https://doi.org/10.3390/ijms21082878.Search in Google Scholar

McKeague, M., Otto, C., Raz, M.H., Angelov, T., and Sturla, S.J. (2018). The base pairing partner modulates alkylguanine alkyltransferase. ACS Chem. Biol. 13: 2534–2541, https://doi.org/10.1021/acschembio.8b00446.Search in Google Scholar

Musarrat, J., Wilson, J.A., Abouissa, H., and Wani, A.A. (1995). O6-alkylguanine DNA alkyltransferase activity levels in normal, benign and malignant human female breast. Biochem. Biophys. Res. Commun. 208: 688–696, https://doi.org/10.1006/bbrc.1995.1393.Search in Google Scholar

Nakatsuru, Y., Matsukuma, S., Nemoto, N., Sugano, H., Sekiguchi, M., and Ishikawa, T. (1993). O6-methylguanine-DNA methyltransferase protects against nitrosamine-induced hepatocarcinogenesis. Proc. Natl. Acad. Sci. U. S. A. 90: 6468–6472, https://doi.org/10.1073/pnas.90.14.6468.Search in Google Scholar

Patra, N., Ioannidis, E.I., and Kulik, H.J. (2016). Computational investigation of the interplay of substrate positioning and reactivity in catechol O-methyltransferase. PloS One 11: e0161868, https://doi.org/10.1371/journal.pone.0161868.8.Search in Google Scholar

Paulsen, M. and Ferguson-Smith, A.C. (2001). DNA methylation in genomic imprinting, development, and disease. J. Pathol. 195: 97–110, https://doi.org/10.1002/path.890.Search in Google Scholar

Perugino, G., Vettone, A., Illiano, G., Valenti, A., Ferrara, M.C., Rossi, M., and Ciaramella, M. (2012). Activity and regulation of archaeal DNA alkyltransferase: conserved protein involved in repair of DNA alkylation damage. J. Biol. Chem. 287: 4222–4231, https://doi.org/10.1074/jbc.m111.308320.Search in Google Scholar

Phillips, J.C., Braun, R., Wang, W., Gumbart, J., Tajkhorshid, E., Villa, E., Chipot, C., Skeel, R.D., Kale, L., and Schulten, K. (2005). Scalable molecular dynamics with NAMD. J. Comput. Chem. 26: 1781–1802, https://doi.org/10.1002/jcc.20289.Search in Google Scholar

Pokhrel, R., Pavadai, E., Gerstman, B.S., and Chapagain, P.P. (2019). Membrane pore formation and ion selectivity of the Ebola virus delta peptide. Phys. Chem. Chem. Phys. 21: 5578–5585, https://doi.org/10.1039/c8cp07323f.Search in Google Scholar

Rasimas, J.J., Pegg, A.E., and Fried, M.G. (2003). DNA-binding mechanism of O6-alkylguanine-DNA alkyltransferase. Effects of protein and DNA alkylation on complex stability. J. Biol. Chem. 278: 7973–7980, https://doi.org/10.1074/jbc.m211854200.Search in Google Scholar

Rossi, F., Morrone, C., Massarotti, A., Ferraris, D.M., Valenti, A., Perugino, G., and Miggiano, R. (2018). Crystal structure of a thermophilic O6-alkylguanine-DNA alkyltransferase-derived self-labeling protein-tag in covalent complex with a fluorescent probe. Biochem. Biophys. Res. Commun. 500: 698–703, https://doi.org/10.1016/j.bbrc.2018.04.139.Search in Google Scholar

Sakumi, K., Shiraishi, A., Shimizu, S., Tsuzuki, T., Ishikawa, T., and Sekiguchi, M. (1997). Methylnitrosourea-induced tumorigenesis in MGMT gene knockout mice. Canc. Res. 57: 2415–2418.Search in Google Scholar

Sugita, Y., Kitao, A., and Okamoto, Y. (2000). Multidimensional replica-exchange method for free-energy calculations. J. Chem. Phys. 113: 6042–6051, https://doi.org/10.1063/1.1308516.Search in Google Scholar

Tessmer, I. and Fried, M.G. (2014). Insight into the cooperative DNA binding of the O(6)-alkylguanine DNA alkyltransferase. DNA Repair 20: 14–22, https://doi.org/10.1016/j.dnarep.2014.01.006.Search in Google Scholar

Tessmer, I., Melikishvili, M., and Fried, M.G. (2012). Cooperative cluster formation, DNA bending and base-flipping by O6-alkylguanine-DNA alkyltransferase. Nucleic Acids Res. 40: 8296–8308, https://doi.org/10.1093/nar/gks574.Search in Google Scholar

Tiwari, P.B., Chapagain, P.P., Banda, S., Darici, Y., Üren, A., and Tse-Dinh, Y.-C. (2016). Characterization of molecular interactions between Escherichia coli RNA polymerase and topoisomerase I by molecular simulations. FEBS Lett. 590: 2844–2851, https://doi.org/10.1002/1873-3468.12321.Search in Google Scholar

Tiwari, P.B., Chapagain, P.P., Seddek, A., Annamalai, T., Üren, A., and Tse-Dinh, Y.-C. (2020). Covalent complex of DNA and bacterial topoisomerase: implications in antibacterial drug development. ChemMedChem 15: 623–631, https://doi.org/10.1002/cmdc.201900721.Search in Google Scholar

Tiwari, P.B., Chapagain, P.P., and Üren, A. (2018). Investigating molecular interactions between oxidized neuroglobin and cytochrome c. Sci. Rep. 8: 10557, https://doi.org/10.1038/s41598-018-28836-6.Search in Google Scholar

Tubbs, J.L., Pegg, A.E., and Tainer, J.A. (2007). DNA binding, nucleotide flipping, and the helix-turn-helix motif in base repair by O6-alkylguanine-DNA alkyltransferase and its implications for cancer chemotherapy. DNA Repair 6: 1100–1115, https://doi.org/10.1016/j.dnarep.2007.03.011.Search in Google Scholar

Warren, J.J., Forsberg, L.J., and Beese, L.S. (2006). The structural basis for the mutagenicity of O6-methyl-guanine lesions. Proc. Natl. Acad. Sci. U. S. A. 103: 19701–19706, https://doi.org/10.1073/pnas.0609580103.Search in Google Scholar

Zak, P., Kleibl, K., and Laval, F. (1994). Repair of O6-methylguanine and O4-methylthymine by the human and rat O6-methylguanine-DNA methyltransferases. J. Biol. Chem. 269: 730–733, https://doi.org/10.1016/s0021-9258(17)42410-0.Search in Google Scholar

Zang, H., Fang, Q., Pegg, A.E., and Guengerich, F.P. (2005). Kinetic analysis of steps in the repair of damaged DNA by human O6-alkylguanine-DNA alkyltransferase. J. Biol. Chem. 280: 30873–30881, https://doi.org/10.1074/jbc.m505283200.Search in Google Scholar


Supplementary Material

The online version of this article offers supplementary material (https://doi.org/10.1515/hsz-2021-0198).


Received: 2021-03-19
Accepted: 2021-05-29
Published Online: 2021-06-30
Published in Print: 2021-09-27

© 2021 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 20.4.2024 from https://www.degruyter.com/document/doi/10.1515/hsz-2021-0198/html
Scroll to top button