Small molecule-mediated allosteric activation of the base excision repair enzyme 8-oxoguanine DNA glycosylase and its impact on mitochondrial function

Gaochao Tian, Steven R Katchur, Yong Jiang, Jacques Briand, Michael Schaber, Constantine Kreatsoulas, Benjamin Schwartz, Sara Thrall, Alicia M Davis, Sam Duvall, Brett A Kaufman, William L Rumsey, Gaochao Tian, Steven R Katchur, Yong Jiang, Jacques Briand, Michael Schaber, Constantine Kreatsoulas, Benjamin Schwartz, Sara Thrall, Alicia M Davis, Sam Duvall, Brett A Kaufman, William L Rumsey

Abstract

8-Oxoguanine DNA glycosylase (OGG1) initiates base excision repair of the oxidative DNA damage product 8-oxoguanine. OGG1 is bifunctional; catalyzing glycosyl bond cleavage, followed by phosphodiester backbone incision via a β-elimination apurinic lyase reaction. The product from the glycosylase reaction, 8-oxoguanine, and its analogues, 8-bromoguanine and 8-aminoguanine, trigger the rate-limiting AP lyase reaction. The precise activation mechanism remains unclear. The product-assisted catalysis hypothesis suggests that 8-oxoguanine and analogues bind at the product recognition (PR) pocket to enhance strand cleavage as catalytic bases. Alternatively, they may allosterically activate OGG1 by binding outside of the PR pocket to induce an active-site conformational change to accelerate apurinic lyase. Herein, steady-state kinetic analyses demonstrated random binding of substrate and activator. 9-Deazaguanine, which can't function as a substrate-competent base, activated OGG1, albeit with a lower Emax value than 8-bromoguanine and 8-aminoguanine. Random compound screening identified small molecules with Emax values similar to 8-bromoguanine. Paraquat-induced mitochondrial dysfunction was attenuated by several small molecule OGG1 activators; benefits included enhanced mitochondrial membrane and DNA integrity, less cytochrome c translocation, ATP preservation, and mitochondrial membrane dynamics. Our results support an allosteric mechanism of OGG1 and not product-assisted catalysis. OGG1 small molecule activators may improve mitochondrial function in oxidative stress-related diseases.

Conflict of interest statement

WLR is chief scientific officer, serves on the board and owns stock in Luciole Pharmaceuticals, Inc, a startup company devoted to development of OGG1 activators. All other authors have no competing interests.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
OGG1 catalytic mechanism. The panels describe the structural interactions between DNA containing 8-oxo-dG and OGG1 represented by the lys249 amino acid. In (a), the glycosylase reaction is displayed and in (b) the AP lyase one with the projected re-arrangements resulting from product assisted catalysis (i) or allosteric modification (ii).
Figure 2
Figure 2
Kinetic mechanisms of enzyme activation. In these kinetic schemes, the symbols represent: E, enzyme, S, substrate, A, activator, Km, Michaelis-Menton constant, Ka, activation constant, α, the coefficient of interaction between activator and substrate, and β, factor of kcat activation. Equations that describe these mechanisms of random binding, ordered with substrate binding first, and ordered with activator binding first are also provided where V is the maximal initial rate (kcatEt, in which Et is the total enzyme concentration). Further details are found in the Supplementary Table S2.
Figure 3
Figure 3
Activation of OGG1 by 8-bromoGua and 9-deazaGua. The term [S] refers to substrate A displayed in Supplementary Fig. S1. Values were obtained in duplicate from an independent experiment by varying substrate concentration over time. In (a) OGG1 (at 10 nM) initial rates were measured as a function of substrate A concentration listed on the x-axis in the presence of 8-bromoGua at 0.0 (×), 0.63 (♢), 2.5 (▽), 5 (△), 10 (◻), and 20 (○) μM. Kinetic parameters from analyzing the data using Eq. (2) were used to calculate the theoretical values (solid lines). (b) Bar plot of OGG1 activity in the presence of 200 μM 8-bromoGua and 9-deazaGua. Data were normalized to OGG1 activity in the presence of 200 μM 8-bromoGua. (c) Similarly, to panel (a), plots of OGG1 (at 10 nM) initial rates as a function of substrate A concentration (x-axis) in the presence of 9-deazaGua at 0.0 (●), 39 (+), 78 (×), 156 (♢), 313 (▽), 625 (△), 1250 (◻), and 2500 (○) μM. (d) Kinetic parameters from analyzing the data using Eq. (2) (Supplementary Table 2) were used to calculate the theoretical values (solid lines). Standard deviation of the fit of the lines provides the error terms.
Figure 4
Figure 4
19F NMR Binding Experiments. (a) Non 1H-decoupled 19F NMR spectra of compound VI in the presence of different concentrations of OGG1. Gradual line broadening of the 19F NMR resonance as OGG1 concentration is increased indicates an interaction with OGG1. (b) Non 1H-decoupled 19F NMR spectra of 100 μM compound VI in the absence of OGG1 (top), presence of 16 μM OGG1 (middle) and presence of 16 μM OGG1 and 100 μM compound VII (bottom). The line narrowing effect observed on the bottom spectrum of Compound VI when compared to the middle spectrum is indicative of the two compounds competing for the same binding site. (c) Bar graph of the average peak heights extracted from the 19F NMR spectra (N = 2) of compound VI obtained with various OGG1 concentrations shown in panel c (dark grey on the left) and in the presence of 16 μM OGG1 plus 100 μM of a displacement compound, VII-X (light grey on the right). For the displacement experiments, the peak height of compound VI can be observed to increase, reflecting a line narrowing of its NMR resonance.
Figure 5
Figure 5
Proposed sites for small molecule OGG1 activator binding. OGG1 (blue ribbon) shown with 8-oxo-dG (yellow spheres) and DNA oligonucleotide (gold) from the cross-liked complex (PDB ID 1HU0). Hot spots (AG) that were identified using mixed probe molecular dynamics were explored via docking experiments. Three hot spots, (B), (E), and (G) could accommodate > 9 small molecule activators.
Figure 6
Figure 6
Effects of paraquat on mitochondrial membrane integrity, mtDNA damage and cytochrome c translocation. In (a) baseline values of mitochondrial integrity are noted by the punctate orange colors whereas in (b), paraquat exposure for 24 h elicits a diffuse characteristic in A549 cells. MitoTracker™ orange dye (overlaid orange) was used to demarcate mitochondria (red arrows) and Hoechst 33342 (blue) to denote the nucleus. (c) provides graphical representation of the mean values obtained from the imaging data from 3 independent experiments. Asterisks represent p < 0.0001 by 1-way ANOVA with Dunnet’s post hoc analyses. In (d), paraquat concentration-dependently damaged mtDNA as measured using two mitochondrial primers described in Methods. Values are depicted as percent of baseline control measurements. For (eg), representative examples obtained from 3 independent experiments show the impact of compounds XI, XII and VII, respectively, on preventing mtDNA damage. For (hj), changes in cytochrome c translocation are depicted in representative images using anti-cytochrome c-DyLight™-550 (yellow), CellMask™ Deep Red (red) and Hoechst 33342. In this case, OGG1 activators were applied 2 h prior to paraquat rather than 4 h in the other experiments. For (i) statistical analyses (ANOVA with Dunnett’s post-test), asterisks represent p < 0.0001 and for (j) PQ value was p < 0.0001 vs control while 30 and 50 µM of the test compound resulted in p = 0.0002 and 0.0003, respectively.
Figure 7
Figure 7
Paraquat-induced changes on mitochondrial dynamics. A549 cells were exposed to paraquat for 48 h and/or pre-treated with OGG1 activators for 4 h. Images were obtained and from these images graphical representations are displayed for (a) TOM20, (b) DRP1 and (c) MFN1. The dotted lines refer to the level of change induced by 0.6 mM paraquat. Values represent the average of two independent experiments in triplicate. Statistical significance was conducted and denoted as described in Fig. 6. For compound VII in (a), p = 0.042. In (b) p values from left to right side of the panel denoted by the asterisks equal 0.0138, 0.0023, 0.0002, < 0.0001, < 0.0001, 0.0242, 0.0002, 0.0003 and < 0.0001, respectively. For (c) the asterisks from left to right of the panel represent p-values of; 0.0099, 0.0498, 0.0183, 0.0045, 0.005 and < 0.0001, respectively.
Figure 8
Figure 8
Effects of paraquat on DNA Ligase III in the presence and absence of OGG1 activators. Panels (ac) represent images obtained in the absence and presence of 0.6 and 1.0 mM paraquat for 48 h, respectively. Sample images depict changes in DNA ligase III content (overlaid orange) with overlaid Cell Mask™ Deep Red (red) and Hoechst 33342 (blue). The line = 40 microns. In (d), graphical representation of the changes resulting in DNA ligase III content after exposure to 0.6 mM paraquat with the dotted line used to depict the changes brought about by the presence of the OGG1 activators (4 h pre-treatment). Similar data were obtained with 24 h of paraquat treatment (data not shown). Values are representative of two independent experiments performed in triplicate. In (d) p-values denoted by the asterisks from left to right of the panel equal 0.0427, 0.0448, 0.0083, 0.0239 and < 0.0001, respectively.

References

    1. Barnes DE, Lindahl T. Repair and genetic consequences of endogenous DNA base damage in mammalian cells. Annu. Rev. Genet. 2004;38:445–476. doi: 10.1146/annurev.genet.38.072902.092448.
    1. Radak Z, Boldogh I. 8-oxo-7,8-dihydroguanine: Link to gene expression, aging and defense against oxidative stress. Free Radic. Biol. Med. 2010;49:587–596. doi: 10.1016/j.freeradbiomed.2010.05.008.
    1. David SS, O’Shea VL, Kundu S. Base excision repair of oxidative DNA damage. Nature. 2007;447:941–950. doi: 10.1038/nature05978.
    1. Peoples MC, Karnes HT. Recent developments in analytical methodology for 8-hydroxy-2′-deoxyguanosine and related compounds. J. Chromatogr. B. 2005;827:5–15. doi: 10.1016/j.jchromb.2005.10.001.
    1. Shibutani S, Takeshita M, Grollman AP. Insertion of specific bases during DNA synthesis past the oxidation-damaged base 8-oxodG. Nature. 1991;394:431–434. doi: 10.1038/349431a0.
    1. Grollman AP, Moriya M. Mutagenesis by 8-oxoguanine: An enemy within. Trends Genet. 1993;9:246–249. doi: 10.1016/0168-9525(93)90089-Z.
    1. Kalyani RR, Corriere M, Ferrucci L. Age-related and disease-related muscle loss: The effect of diabetes, obesity, and other diseases. Lancet Diabetes Endocrinol. 2014;2:819–829. doi: 10.1016/S2213-8587(14)70034-8.
    1. Sheng Z, et al. 8-Oxoguanine causes neurodegeneration during MUTYH-mediated DNA base excision repair. J. Clin. Investig. 2012;122:4344–4361. doi: 10.1172/JCI65053.
    1. Pastukh VM, et al. Oxidative DNA damage in lung tissue from patients with COPD is clustered in functionally significant sequences. Int. J. Chronic Obstr. Pulm. Dis. 2011;6:209–217.
    1. Cooke MS, Evans MD, Dizdaroglu M, Lunec J. Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J. 2003;17:1195–1214. doi: 10.1096/fj.02-0752rev.
    1. Simone S, Gorin Y, Velagapudi C, Abboud HE, Habib SL. Mechanism of oxidative DNA damage in diabetes-tuberin inactivation and downregulation of DNA repair enzyme 8-oxo-7,8-dihydro-2-deoxyguanosine-DNA glycosylase. Diabetes. 2008;57:2626–2636. doi: 10.2337/db07-1579.
    1. Ide T. Mitochondrial DNA damage and dysfunction associated with oxidative stress in failing hearts after myocardial infarction. Circ. Res. 2001;88:529–535. doi: 10.1161/01.RES.88.5.529.
    1. Vlahopoulos S, Adamaki M, Khoury N, Zoumpourlis V, Boldogh I. Roles of DNA repair enzyme OGG1 in innate immunity and its significance for lung cancer. Pharmacol. Ther. 2018;194:59–72. doi: 10.1016/j.pharmthera.2018.09.004.
    1. Krokan HE, Bjørås M. Base excision repair. Cold Spring Harb. Perspect. Biol. 2013;5:1–22. doi: 10.1101/cshperspect.a012583.
    1. Zharkov DO. Base excision DNA repair. Cell. Mol. Life Sci. 2008;65:1544–1565. doi: 10.1007/s00018-008-7543-2.
    1. Robertson AB, Klungland A, Rognes T, Leiros I. Base excision repair: The long and short of it. Cell. Mol. Life Sci. 2009;66:981–993. doi: 10.1007/s00018-009-8736-z.
    1. Ruchko MV, Gorodnya OM, Zuleta A, Pastukh VM, Gillespie MN. The DNA glycosylase, Ogg1, defends against oxidant-induced mtDNA damage and apoptosis in pulmonary artery endothelial cells. Free Radic. Biol. Med. 2011;50:1107–1113. doi: 10.1016/j.freeradbiomed.2010.10.692.
    1. Komakula SSB, et al. The DNA repair protein OGG1 protects against obesity by altering mitochondrial energetics in white adipose tissue. Sci. Rep. 2018;8:14886. doi: 10.1038/s41598-018-33151-1.
    1. Tan YB, et al. Pharmacological Protection of mitochondrial DNA integrity may afford a new strategy for suppressing lung ischemia-reperfusion injury. Ann. Am. Thorac. Soc. 2017;14(3):S210–S215. doi: 10.1513/AnnalsATS.201706-438MG.
    1. Torres-Gonzalez M, Gawlowski T, Kocalis H, Scott BT, Dillmann WH. Mitochondrial 8-oxoguanine glycosylase decreases mitochondrial fragmentation and improves mitochondrial function in H9C2 cells under oxidative stress conditions. Am. J. Physiol. Cell Physiol. 2014;306:C221–C229. doi: 10.1152/ajpcell.00140.2013.
    1. Yuzefovych LV, et al. Alteration of mitochondrial function and insulin sensitivity in primary mouse skeletal muscle cells isolated from transgenic and knockout mice: Role of OGG1. Endocrinology. 2013;154(8):2640–2649. doi: 10.1210/en.2013-1076.
    1. Nash HM, Lu L, Lane WS, Verdine GL. The critical active-site amine of the human 8-oxoguanine DNA glycosylase, hOgg1: Direct identification, ablation and chemical reconstitution. Chem. Biol. 1997;4:693–702. doi: 10.1016/S1074-5521(97)90225-8.
    1. Tyugshev TE, Vorobjev YN, Kuznetsova AA, Lukina MV, Kuznetsov NA, Fedorova OS. Roles of active-site amino acid residues in specific recognition of DNA lesions by human 8-oxoguanine-DNA glycosylase (OGG1) J. Phys. Chem. B. 2019;123:4878–4887. doi: 10.1021/acs.jpcb.9b02949.
    1. Fromme JC, Bruner SD, Yang W, Karplus M, Verdine GL. Product-assisted catalysis in base-excision DNA repair. Nature Struct. Biol. 2003;10:204–211. doi: 10.1038/nsb902.
    1. Kuznetsov NA, Koval VV, Zharkov DO, Nevinsky GA, Douglas KT, Fedorova OS. Kinetics of substrate recognition and cleavage by human 8-oxoguanine-DNA glycosylase. Nucl. Acids Res. 2005;33(12):3919–3931. doi: 10.1093/nar/gki694.
    1. Boldogh I, et al. Activation of ras signaling pathway by 8-oxoguanine DNA glycosylase bound to its excision product, 8-oxoguanine. J. Biol. Chem. 2012;287:20769–20773. doi: 10.1074/jbc.C112.364620.
    1. Baptiste BA, et al. Enhanced mitochondrial DNA repair of the common disease-associated variant, Ser326Cys, of hOGG1 through small molecule intervention. Free Radic. Biol. Med. 2018;124:149–162. doi: 10.1016/j.freeradbiomed.2018.05.094.
    1. Mirbahai L, et al. Use of a molecular beacon to track the activity of base excision repair protein OGG1 in live cells. DNA Repair. 2010;9:144–152. doi: 10.1016/j.dnarep.2009.11.009.
    1. Banerjee A, Yang W, Karplus M, Verdine GL. Structure of a repair enzyme interrogating undamaged DNA elucidates recognition of damaged DNA. Nature. 2005;434:612–618. doi: 10.1038/nature03458.
    1. Cocheme HM, Murphy MP. Complex I is the major site of mitochondrial superoxide production by paraquat. J. Biol. Chem. 2008;283(4):1786–1798. doi: 10.1074/jbc.M708597200.
    1. Dwyer Z, et al. Characterizing the protracted neurobiological and neuroanatomical effects of paraquat in a murine model of Parkinson's disease. Neurobiol. Aging. 2021;100:11–21. doi: 10.1016/j.neurobiolaging.2020.11.013.
    1. Subbiah R, Tiwari RR. The herbicide paraquat-induced molecular mechanisms in the development of acute lung injury and lung fibrosis. Crit. Rev. Toxicol. 2021;51(1):36–64. doi: 10.1080/10408444.2020.1864721.
    1. West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat. Rev. Immunol. 2017;17(6):363–375. doi: 10.1038/nri.2017.21.
    1. Furda A, Marrangoni AM, Lokshin A, Van Houten B. Oxidants and not alkylating agents induce rapid mtDNA loss and mitochondrial dysfunction. DNA Repair (Amst) 2012;11:684–692. doi: 10.1016/j.dnarep.2012.06.002.
    1. Chen N, et al. Drp1-mediated mitochondrial fission contributes to mitophagy in paraquat-induced neuronal cell damage. Environ. Pollut. 2021;272:116413–116426. doi: 10.1016/j.envpol.2020.116413.
    1. Akbari M, et al. Overexpression of DNA ligase III in mitochondria protects cells against oxidative stress and improves mitochondrial DNA base excision repair. DNA Repair (Amst) 2014;16:44–53. doi: 10.1016/j.dnarep.2014.01.015.
    1. Kobayashi TJ, Yokota R, Aihara K. Feedback regulation and Its efficiency in biochemical networks. J. Stat. Phys. 2016;162:1425–1449. doi: 10.1007/s10955-015-1443-2.
    1. Dalhus B, et al. Separation-of-function mutants unravel the dual-reaction mode of human 8-oxoguanine dna glycosylase. Structure. 2011;19:117–127. doi: 10.1016/j.str.2010.09.023.
    1. Liu D, et al. Evidence that OGG1 glycosylase protects neurons against oxidative DNA damage and cell death under ischemic conditions. J. Cerebr. Blood Flow Metab. 2011;31:680–692. doi: 10.1038/jcbfm.2010.147.
    1. Kuznetsov NA, Koval VV, Nevinsky GA, Douglas KT, Zharkov DO, Fedorova OS. Kinetic conformational analysis of human 8-oxoguanine-DNA glycosylase. J. Biol. Chem. 2007;282:1029–1038. doi: 10.1074/jbc.M605788200.
    1. Norman DPG, Chung SJ, Verdine GL. Structural and biochemical exploration of a critical amino acid in human 8-oxoguanine glycosylase. Biochemistry. 2003;42:1564–1572. doi: 10.1021/bi026823d.
    1. Riley JS, Tait SWG. Mitochondrial inflammation and immunity. EMBO Rep. 2020;21:e49799. doi: 10.15252/embr.201949799.
    1. Leon J, Sakumi K, Castillo E, Sheng Z, Oka S, Nakabeppu Y. 8-Oxoguanine accumulation in mitochondrial DNA causes mitochondrial dysfunction and impairs neuritogenesis in cultured adult mouse cortical neurons under oxidative conditions. Sci. Rep. 2016;6:22086. doi: 10.1038/srep22086.
    1. Lia D, et al. Mitochondrial maintenance under oxidative stress depends on mitochondrially localised α-OGG1. J. Cell Sci. 2018;131:jcs213538. doi: 10.1242/jcs.213538.
    1. Stuart JA, Bourque BM, de Souza-Pinto NC, Bohr VA. No evidence of mitochondrial respiratory dysfunction in OGG1-null mice deficient in removal of 8-oxodeoxyguanine from mitochondrial DNA. Free Radic. Biol. Med. 2005;38:737–745. doi: 10.1016/j.freeradbiomed.2004.12.003.
    1. Oka S, Ohno M, Tsuchimoto D, Sakumi K, Furuichi M, Nakabeppu Y. Two distinct pathways of cell death triggered by oxidative damage to nuclear and mitochondrial DNAs. EMBO J. 2008;27:421–432. doi: 10.1038/sj.emboj.7601975.
    1. Dickinson A, et al. The regulation of mitochondrial DNA copy number in glioblastoma cells. Cell Death Differ. 2013;20:1644–1653. doi: 10.1038/cdd.2013.115.
    1. Pinz KG, Shibutani S, Bogenhagen DF. Action of mitochondrial DNA Polymerase g at sites of base loss or oxidative damage. J. Biol. Chem. 1995;270(16):9202–9206. doi: 10.1074/jbc.270.16.9202.
    1. Filograna R, Mennuni M, Alsina D, Larsson NG. Mitochondrial copy number in human disease: The more the better? Febs Lett. 2021;595:976–1002. doi: 10.1002/1873-3468.14021.
    1. Filograna R, et al. Modulation of mtDNA copy number ameliorates the pathological consequences of a heteroplasmic mtDNA mutation in the mouse. Sci. Adv. 2019;5:1–13. doi: 10.1126/sciadv.aav9824.
    1. Tian Q, et al. Mitochondrial DNA copy number and heteroplasmy load correlate with skeletal muscle oxidative capacity by P31 MR spectroscopy. Aging Cell. 2021 doi: 10.1111/acel.13487.
    1. de Souza-Pinto NC, et al. Repair of 8-oxodeoxyguanosine lesions in mitochondrial DNA depends on the oxoguanine DNA glycosylase (OGG1) gene and 8-oxoguanine accumulates in the mitochondrial DNA of OGG1-defective mice. Cancer Res. 2001;61:5378–5381.
    1. Cardozo-Pelaez F, Sanchez-Contreras M, Nevin ABC. Ogg1 null mice exhibit age-associated loss of the nigrostriatal pathway and increased sensitivity to MPTP. Neurochem. Int. 2012;61(5):721–730. doi: 10.1016/j.neuint.2012.06.013.
    1. Sampath H, Vartanian V, Rollins MR, Sakumi K, Nakabeppu Y, Lloyd S. 8-Oxoguanine DNA Glycosylase (OGG1) deficiency increases susceptibility to obesity and metabolic dysfunction. PLoS ONE. 2012;7(12):e51697. doi: 10.1371/journal.pone.0051697.
    1. Dincer Y, et al. DNA repair gene OGG1 polymorphism and its relation with oxidative DNA damage in patients with Alzheimer’s disease. Neurosci. Lett. 2019 doi: 10.1016/j.neulet.2019.134362.
    1. Corella D, et al. Effects of the Ser326Cys Polymorphism in the DNA Repair OGG1 Gene on Cancer, Cardiovascular, and all-cause mortality in the predimed study: Modulation by diet. J. Acad. Nutr. Diet. 2018;118(4):589–605. doi: 10.1016/j.jand.2017.09.025.
    1. Bravard A, et al. Redox Regulation of Human OGG1 Activity in Response to Cellular Oxidative Stress. Mol. Cell. Biol. 2006;26(20):7430–7436. doi: 10.1128/MCB.00624-06.
    1. Bravard A, et al. Oxidation status of human OGG1-S326C polymorphic variant determines cellular DNA repair capacity. Cancer Res. 2009;69(8):3642–3649. doi: 10.1158/0008-5472.CAN-08-3943.
    1. Michel M, et al. Small-molecule activation of OGG1 increases oxidative DNA damage repair by gaining a new function. Science. 2022;376:1471–1476. doi: 10.1126/science.abf8980.
    1. Mabley JG, et al. Potential role for 8-oxoguanine DNA glycosylase in regulating inflammation. FASEB J. 2004 doi: 10.1096/fj.04-2278fje.
    1. Visnes T, et al. Small-molecule inhibitor of OGG1 suppresses proinflammatory gene expression and inflammation. Science. 2018;362:834–839. doi: 10.1126/science.aar8048.
    1. Hanna Bishoy M. F., Helleday Thomas, Mortusewicz Oliver. OGG1 Inhibitor TH5487 Alters OGG1 Chromatin Dynamics and Prevents Incisions. Biomolecules. 2020;10(11):1483. doi: 10.3390/biom10111483.
    1. Xian H, et al. Oxidized DNA fragments exit mitochondria via mPTP- and VDAC-dependent channels to activate NLRP3 inflammasome and interferon signaling. Immunity. 2022;55:1–16. doi: 10.1016/j.immuni.2022.06.007.
    1. Panduri V, et al. Role of mitochondrial hOGG1 and aconitase in oxidant-induced lung epithelial cell apoptosis. Free Radic. Biol. Med. 2009;47(6):750–759. doi: 10.1016/j.freeradbiomed.2009.06.010.
    1. Bakan A, Nevins N, Lakdawala AS, Bahar I. Druggability assessment of allosteric proteins by dynamics simulations in the presence of probe molecules. J. Chem. Theory Comput. 2012;8:2435–2447. doi: 10.1021/ct300117j.
    1. Bjoras M, Seeberg E, Luna L, Pearl LH, Barrett TE. Reciprocal "flipping" underlies substrate recognition and catalytic activation by the human 8-oxo-guanine DNA glycosylase. J. Mol. Biol. 2002;317:171–177. doi: 10.1006/jmbi.2002.5400.
    1. Friesner RA, et al. Extra precision glide: Docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 2006;49:6177–6196. doi: 10.1021/jm051256o.
    1. Banks JL, et al. Integrated modeling program, applied chemical theory (IMPACT) J. Comp. Chem. 2005;26:1752–1780. doi: 10.1002/jcc.20292.
    1. Jacobson M, et al. Hierarchical approach to all-atom protein loop prediction. Protein Struct. Funct. Bioinform. 2004;55:351–367. doi: 10.1002/prot.10613.
    1. Kolesar J.E. & Kaufman B.A. Using two-dimensional intact mitochondrial DNA (mtDNA) agarose gel electrophoresis (2D-IMAGE) to detect changes in topology associated with mitochondrial replication, transcription, and damage. in Hanada K. (ed)DNA Electrophoresis. Methods in Molecular Biology211910.1007/978-1-0716-0323-9_3 (Humana, 2020).
    1. Falabella M, et al. G-quadruplex dynamics contribute to regulation of mitochondrial gene expression. Sci. Rep. 2019;9(1):5605–5622. doi: 10.1038/s41598-019-41464-y.
    1. Furda, A., Santos, J.H., Meyer, J.N. & Van Houten, B. Quantitative PCR-Based Measurement of nuclear and mitochondrial DNA damage and repair in mammalian cells. in Keohavong P., & Grant S. (eds.) Molecular Toxicology Protocols. Methods in Molecular Biology (Methods and Protocols), 1105,10.1007/978-1-62703-739-6_31 (Humana Press, 2014).
    1. Dai Y, Zheng K, Clark J, Swerdlow RH. Rapamycin drives selection against a pathogenic heteroplasmic mitochondrial DNA mutation. Hum. Genet. 2014;23:637–647.
    1. Mortusewicz O, Rothbauer U, Cardoso MC, Leonhardt H. Differential recruitment of DNA ligase to DNA repair sites. Nucl. Acids Res. 2006;34:3523–3532. doi: 10.1093/nar/gkl492.

Source: PubMed

3
Subscribe