Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Oxidative guanine base damage regulates human telomerase activity

This article has been updated

Abstract

Changes in telomere length are associated with degenerative diseases and cancer. Oxidative stress and DNA damage have been linked to both positive and negative alterations in telomere length and integrity. Here we examined how the common oxidative lesion 8-oxo-7,8-dihydro-2′-deoxyguanine (8-oxoG) regulates telomere elongation by human telomerase. When 8-oxoG is present in the dNTP pool as 8-oxodGTP, telomerase utilization of the oxidized nucleotide during telomere extension is mutagenic and terminates further elongation. Depletion of MTH1, the enzyme that removes oxidized dNTPs, increases telomere dysfunction and cell death in telomerase-positive cancer cells with shortened telomeres. In contrast, a preexisting 8-oxoG within the telomeric DNA sequence promotes telomerase activity by destabilizing the G-quadruplex DNA structure. We show that the mechanism by which 8-oxoG arises in telomeres, either by insertion of oxidized nucleotides or by direct reaction with free radicals, dictates whether telomerase is inhibited or stimulated and thereby mediates the biological outcome.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: 8-oxodGTP is a telomerase chain terminator.
Figure 2: Telomerase preferentially incorporates 8-oxodGTP opposite rA.
Figure 3: Cells with shortened telomeres are hypersensitive to oxidized dNTPs.
Figure 4: Oxidized dNTPs induce telomere defects in cells with shortened telomeres.
Figure 5: Telomerase elongation of primers with a terminal 8-oxoG.
Figure 6: 8-oxoG restores telomerase activity on quadruplex folded overhangs.
Figure 7: 8-oxoG increases the dynamics of telomeric GQ structures.

Similar content being viewed by others

Change history

  • 14 December 2016

    In the version of this article originally published, the Online Methods section erroneously identified the probe used for immunofluorescence and in situ hybridization assays. The error has been corrected for the PDF and HTML versions of this article.

References

  1. Jaskelioff, M. et al. Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature 469, 102–106 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Armanios, M. & Blackburn, E.H. The telomere syndromes. Nat. Rev. Genet. 13, 693–704 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Greider, C.W. & Blackburn, E.H. A telomeric sequence in the RNA of Tetrahymena telomerase required for telomere repeat synthesis. Nature 337, 331–337 (1989).

    Article  CAS  PubMed  Google Scholar 

  4. Hockemeyer, D. & Collins, K. Control of telomerase action at human telomeres. Nat. Struct. Mol. Biol. 22, 848–852 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kim, N.W. et al. Specific association of human telomerase activity with immortal cells and cancer. Science 266, 2011–2015 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. von Zglinicki, T. Oxidative stress shortens telomeres. Trends Biochem. Sci. 27, 339–344 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Jurk, D. et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2, 4172 (2014).

    Article  PubMed  CAS  Google Scholar 

  8. Lonkar, P. & Dedon, P.C. Reactive species and DNA damage in chronic inflammation: reconciling chemical mechanisms and biological fates. Int. J. Cancer 128, 1999–2009 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Poljšak, B. & Fink, R. The protective role of antioxidants in the defence against ROS/RNS-mediated environmental pollution. Oxid. Med. Cell. Longev. 2014, 671539 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Wang, Z. et al. Characterization of oxidative guanine damage and repair in mammalian telomeres. PLoS Genet. 6, e1000951 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Kamiya, H. & Kasai, H. Formation of 2-hydroxydeoxyadenosine triphosphate, an oxidatively damaged nucleotide, and its incorporation by DNA polymerases: steady-state kinetics of the incorporation. J. Biol. Chem. 270, 19446–19450 (1995).

    Article  CAS  PubMed  Google Scholar 

  12. Freudenthal, B.D. et al. Uncovering the polymerase-induced cytotoxicity of an oxidized nucleotide. Nature 517, 635–639 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Tsuzuki, T. et al. Spontaneous tumorigenesis in mice defective in the MTH1 gene encoding 8-oxo-dGTPase. Proc. Natl. Acad. Sci. USA 98, 11456–11461 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gad, H. et al. MTH1 inhibition eradicates cancer by preventing sanitation of the dNTP pool. Nature 508, 215–221 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Sakumi, K. et al. Cloning and expression of cDNA for a human enzyme that hydrolyzes 8-oxo-dGTP, a mutagenic substrate for DNA synthesis. J. Biol. Chem. 268, 23524–23530 (1993).

    Article  CAS  PubMed  Google Scholar 

  16. Speina, E. et al. Contribution of hMTH1 to the maintenance of 8-oxoguanine levels in lung DNA of non-small-cell lung cancer patients. J. Natl. Cancer Inst. 97, 384–395 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Patel, A. et al. MutT Homolog 1 (MTH1) maintains multiple KRAS-driven pro-malignant pathways. Oncogene 34, 2586–2596 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Liou, G.Y. & Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 44, 479–496 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Szatrowski, T.P. & Nathan, C.F. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res. 51, 794–798 (1991).

    CAS  PubMed  Google Scholar 

  20. Wallace, S.S. Base excision repair: a critical player in many games. DNA Repair (Amst.) 19, 14–26 (2014).

    Article  CAS  Google Scholar 

  21. Zhou, J., Liu, M., Fleming, A.M., Burrows, C.J. & Wallace, S.S. Neil3 and NEIL1 DNA glycosylases remove oxidative damages from quadruplex DNA and exhibit preferences for lesions in the telomeric sequence context. J. Biol. Chem. 288, 27263–27272 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Askree, S.H. et al. A genome-wide screen for Saccharomyces cerevisiae deletion mutants that affect telomere length. Proc. Natl. Acad. Sci. USA 101, 8658–8663 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lu, J. & Liu, Y. Deletion of Ogg1 DNA glycosylase results in telomere base damage and length alteration in yeast. EMBO J 29, 398–409 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Schmidt, J.C. & Cech, T.R. Human telomerase: biogenesis, trafficking, recruitment, and activation. Genes Dev. 29, 1095–1105 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xi, L. & Cech, T.R. Inventory of telomerase components in human cells reveals multiple subpopulations of hTR and hTERT. Nucleic Acids Res. 42, 8565–8577 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Traut, T.W. Physiological concentrations of purines and pyrimidines. Mol. Cell. Biochem. 140, 1–22 (1994).

    Article  CAS  PubMed  Google Scholar 

  27. Sohl, C.D., Ray, S. & Sweasy, J.B. Pools and Pols: mechanism of a mutator phenotype. Proc. Natl. Acad. Sci. USA 112, 5864–5865 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Katafuchi, A. & Nohmi, T. DNA polymerases involved in the incorporation of oxidized nucleotides into DNA: their efficiency and template base preference. Mutat. Res. 703, 24–31 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Hsu, G.W., Ober, M., Carell, T. & Beese, L.S. Error-prone replication of oxidatively damaged DNA by a high-fidelity DNA polymerase. Nature 431, 217–221 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Bertram, J.G., Oertell, K., Petruska, J. & Goodman, M.F. DNA polymerase fidelity: comparing direct competition of right and wrong dNTP substrates with steady state and pre-steady state kinetics. Biochemistry 49, 20–28 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Tomlinson, C.G. et al. Two-step mechanism involving active-site conformational changes regulates human telomerase DNA binding. Biochem. J. 465, 347–357 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Rai, P. et al. Continuous elimination of oxidized nucleotides is necessary to prevent rapid onset of cellular senescence. Proc. Natl. Acad. Sci. USA 106, 169–174 (2009).

    Article  PubMed  Google Scholar 

  33. Crabbe, L., Cesare, A.J., Kasuboski, J.M., Fitzpatrick, J.A. & Karlseder, J. Human telomeres are tethered to the nuclear envelope during postmitotic nuclear assembly. Cell Rep. 2, 1521–1529 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. O'Sullivan, R.J. et al. Rapid induction of alternative lengthening of telomeres by depletion of the histone chaperone ASF1. Nat. Struct. Mol. Biol. 21, 167–174 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Takai, H., Smogorzewska, A. & de Lange, T. DNA damage foci at dysfunctional telomeres. Curr. Biol. 13, 1549–1556 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Takai, K.K., Hooper, S., Blackwood, S., Gandhi, R. & de Lange, T. In vivo stoichiometry of shelterin components. J. Biol. Chem. 285, 1457–1467 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Sfeir, A. et al. Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell 138, 90–103 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Biffi, G., Tannahill, D., McCafferty, J. & Balasubramanian, S. Quantitative visualization of DNA G-quadruplex structures in human cells. Nat. Chem. 5, 182–186 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hwang, H. et al. Telomeric overhang length determines structural dynamics and accessibility to telomerase and ALT-associated proteins. Structure 22, 842–853 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zahler, A.M., Williamson, J.R., Cech, T.R. & Prescott, D.M. Inhibition of telomerase by G-quartet DNA structures. Nature 350, 718–720 (1991).

    Article  CAS  PubMed  Google Scholar 

  41. Cookson, J.C. et al. Pharmacodynamics of the G-quadruplex-stabilizing telomerase inhibitor 3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]acridinium methosulfate (RHPS4) in vitro: activity in human tumor cells correlates with telomere length and can be enhanced, or antagonized, with cytotoxic agents. Mol. Pharmacol. 68, 1551–1558 (2005).

    Article  CAS  PubMed  Google Scholar 

  42. Meyer, P.R., Matsuura, S.E., So, A.G. & Scott, W.A. Unblocking of chain-terminated primer by HIV-1 reverse transcriptase through a nucleotide-dependent mechanism. Proc. Natl. Acad. Sci. USA 95, 13471–13476 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Vorlícková, M., Tomasko, M., Sagi, A.J., Bednarova, K. & Sagi, J. 8-oxoguanine in a quadruplex of the human telomere DNA sequence. FEBS J. 279, 29–39 (2012).

    Article  PubMed  CAS  Google Scholar 

  44. Tippana, R., Xiao, W. & Myong, S. G-quadruplex conformation and dynamics are determined by loop length and sequence. Nucleic Acids Res. 42, 8106–8114 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Oikawa, S., Tada-Oikawa, S. & Kawanishi, S. Site-specific DNA damage at the GGG sequence by UVA involves acceleration of telomere shortening. Biochemistry 40, 4763–4768 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Hukezalie, K.R., Thumati, N.R., Côté, H.C. & Wong, J.M. In vitro and ex vivo inhibition of human telomerase by anti-HIV nucleoside reverse transcriptase inhibitors (NRTIs) but not by non-NRTIs. PLoS One 7, e47505 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Oulton, R. & Harrington, L. A human telomerase-associated nuclease. Mol. Biol. Cell 15, 3244–3256 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Huard, S. & Autexier, C. Human telomerase catalyzes nucleolytic primer cleavage. Nucleic Acids Res. 32, 2171–2180 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pursell, Z.F., McDonald, J.T., Mathews, C.K. & Kunkel, T.A. Trace amounts of 8-oxo-dGTP in mitochondrial dNTP pools reduce DNA polymerase gamma replication fidelity. Nucleic Acids Res. 36, 2174–2181 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kunkel, T.A. DNA replication fidelity. J. Biol. Chem. 279, 16895–16898 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Kreiter, M., Irion, V., Ward, J. & Morin, G. The fidelity of human telomerase. Nucleic Acids Symp. Ser. 137–139 (1995).

  52. Bisoffi, M., Heaphy, C.M. & Griffith, J.K. Telomeres: prognostic markers for solid tumors. Int. J. Cancer 119, 2255–2260 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Kettle, J.G. et al. Potent and selective inhibitors of MTH1 probe its role in cancer cell survival. J. Med. Chem. 59, 2346–2361 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Zhang, X., Mar, V., Zhou, W., Harrington, L. & Robinson, M.O. Telomere shortening and apoptosis in telomerase-inhibited human tumor cells. Genes Dev. 13, 2388–2399 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mender, I., Gryaznov, S., Dikmen, Z.G., Wright, W.E. & Shay, J.W. Induction of telomere dysfunction mediated by the telomerase substrate precursor 6-thio-2′-deoxyguanosine. Cancer Discov. 5, 82–95 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Rhee, D.B., Ghosh, A., Lu, J., Bohr, V.A. & Liu, Y. Factors that influence telomeric oxidative base damage and repair by DNA glycosylase OGG1. DNA Repair (Amst) 10, 34–44 (2011).

    Article  CAS  Google Scholar 

  57. Giribaldi, M.G., Munoz, A., Halvorsen, K., Patel, A. & Rai, P. MTH1 expression is required for effective transformation by oncogenic HRAS. Oncotarget 6, 11519–11529 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Piskounova, E. et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Fouquerel, E. et al. ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ depletion. Cell Rep. 8, 1819–1831 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Latrick, C.M. & Cech, T.R. POT1-TPP1 enhances telomerase processivity by slowing primer dissociation and aiding translocation. EMBO J 29, 924–933 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pham, H.H. et al. Cooperative hybridization of γPNA miniprobes to a repeating sequence motif and application to telomere analysis. Org. Biomol. Chem. 12, 7345–7354 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ohno, M., Oka, S. & Nakabeppu, Y. Quantitative analysis of oxidized guanine, 8-oxoguanine, in mitochondrial DNA by immunofluorescence method. Methods Mol. Biol. 554, 199–212 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Parikh, D., Fouquerel, E., Murphy, C.T., Wang, H. & Opresko, P.L. Telomeres are partly shielded from ultraviolet-induced damage and proficient for nucleotide excision repair of photoproducts. Nat. Commun. 6, 8214 (2015).

    Article  PubMed  Google Scholar 

  64. Herbert, B.S., Shay, J.W. & Wright, W.E. Analysis of telomeres and telomerase. Curr. Protoc. Cell Biol. 20, 18.16 (2003).

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Watkins and C. St. Croix (University of Pittsburgh Center for Biological Imaging) for assistance with fluorescence imaging, S. Uttam for help with statistical analyses, T. Moiseeva for assistance with the annexin V and propidium iodide FACS assay and R. O'Sullivan and B. Van Houten for critical reading of the manuscript. We also thank T. Cech and A. Zaug (University of Colorado) for reagents and assistance with the telomerase assays and R. O'Sullivan (University of Pittsburgh) for generously providing the HeLa VST and HeLa LT cell lines. This work was supported by NIH grants R01ES022944, R21AG045545 and R21ES025606 (to P.L.O.), American Cancer Society RSG-12-066-01-DMC and NIH 1DP2GM105453 (to T.L., G.K. and S.M.), NIH grant R00ES024431 (to B.D.F.), and NIH grant CA148629 and the Abraham A. Mitchell Distinguished Investigator fund (to R.W.S.). This project used the UPCI CTIF and CF, which are supported in part by award P30CA047904.

Author information

Authors and Affiliations

Authors

Contributions

E.F., J. Lormand and A.B. performed biochemical and cellular experiments, analyzed the data and prepared figures. H.-T.L. and G.S.K. performed all the smFRET studies. J. Li and R.W.S. provided lentiviruses for MTH1 depletion experiments, and R.W.S. provided helpful discussions. E.F., B.D.F., S.M. and P.L.O. designed experiments, analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Patricia L Opresko.

Ethics declarations

Competing interests

R.W.S. is a scientific consultant for Trevigen, Inc. The other authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 8-oxodGTP inhibits telomerase activity.

(a) Telomerase reactions were conducted using (TTAGGG)3 substrate (oligo #1 in Supplementary Table 1) and contained high dNTPs (lanes 1 - 4) or cellular dNTPs (lanes 5 - 8) and 0.3 μM 32P-α-dGTP. Natural dGTP and 8-oxodGTP were mixed at 1:0, 1:1, and 0:1 ratios to maintain a total concentration of 500 μM or 5.2 μM of unlabeled guanine nucleotide. Lanes marked with 0* contained 0.3 μM 32P-α-dGTP, and lacked unlabeled dGTP. Products were separated on polyacrylamide denaturing gels. The LC was a 32P end-labeled 18-mer oligonucleotide. Numbers with plus sign indicate number of telomeric repeats added. (b) Products were quantitated and normalized to the LC, and used to calculate processivity (top graph) and relative activity (bottom graph). Bars represent the mean and s.d. from three independent reactions. Asterisks, ** p < 0.01, *** p < 0.001 based on two-tailed Student’s t test.

Supplementary Figure 2 Elongation termination after 8-oxodGTP is not due to inhibition of telomerase translocation.

Telomerase reactions were conducted using (GGTTAG)3 (oligonucleotide 2 in Supplementary Table 1), and contained cellular dNTPs (37 μM dTTP, 24 μM dATP, 29 μM dCTP, 5.2 μM dGTP) and 0.3 μM 32P-α-dTTP. dGTP and 8-oxodGTP were mixed at 1:0, 1:1, and 0:1 ratios to maintain a total concentration of 5.2 μM of unlabeled guanine nucleotide. Products were separated on polyacrylamide denaturing gels. The LC was a 32P end-labeled 18-mer oligonucleotide. Numbers with plus sign indicate number of repeats added.

Supplementary Figure 3 MTH1 depletion generates DNA damage in HeLa cell lines with very short or long telomeres.

(a) Telomere lengths were confirmed by Southern blotting of telomeric restrictions fragments. The DNA ladder is shown. UD indicates undigested genomic DNA and D indicates digested genomic DNA. (b) Cell lysate was immuno-stained for catalase and SOD expression prior to lentiviral infection. (c) Cells were immuno-stained with an 8-oxoG antibody (green) three days after infection with lentivirus expressing a non-targeting shRNA (scr) or two different shRNAs targeting MTH1 (sh4 or sh5). Nuclei are indicated by DAPI staining (blue). Exposure times and image settings were equivalent for the sh4 and sh5, compared to the scr control. (d) Cells were stained for telomeric DNA by FISH (green) and immuno-stained with a TRF2 antibody (red) three days after lentiviral infection. Nuclei are indicated by DAPI. (e) The average number of TIFs per cell nuclei was calculated after three days post infection with lentivirus. Bars represent the mean ± sd from 3 independent experiments (100 – 150 cells per condition). * p<0.01 based on One-way ANOVA with Tukey’s honest significance difference. (f) Prolong cell culturing partly restores MTH1 expression. Cell lysate was immuno-stained for MTH1 following 138 days after lentiviral infection, which corresponds to population doubling (PD) numbers or HeLa VST of 151 (scr and sh5) or 139 (sh4) and for HeLa LT of 140 (scr and sh5) and 135 (sh4).

Supplementary Figure 4 Detection of cell death in MTH1-depleted cells.

(a and b) Apoptosis was measured by annexin V staining and flow cytometry in HeLa VST or HeLa LT 6 (a) and 9 (b) days after infection with lentivirus expressing a non-targeting shRNA (scr) or different shRNAs against MTH1 (sh4 and sh5). Numbers above red box indicate total annexin V positive cells. Bar graphs show the mean ± s.d. of 4 replicates. ****p<0.0001, by two-tailed Student’s t test. By 9 days post-infection no significant increase in apoptosis is detected in either cell line. (c) MTH1 depletion triggers minimal apoptosis in BJ-hTERT and BJ fibroblasts compared to controls, 3 days after lentivirus infection, compared to controls, as measured by annexin V by flow cytometry.

Supplementary Figure 5 Telomerase activity modulates sensitivity to MTH1 depletion

(a) Immunoblot with antibodies against MTH1 or actin. The percent MTH1 expression relative to the controls are shown. (b) Cells were immuno-stained with an 8oxoG antibody (green) 3 days after infection with lentivirus expressing a non-targeting shRNA (scr) or two different shRNAs targeting MTH1 (sh4 or sh5). Nuclei are indicated by DAPI staining (blue). Exposure times and image settings were equivalent for the sh4 and sh5, compared to the scr control. (c) BJhTERT and BJ cells were fixed 3 days after lentiviral infection and stained for SA-beta-gal activity. The percentage of positive cells is indicated on the bar graph. (d) Whole cell lysates (10μg) were immunoblotted with anti-p53 and anti-actin antibody as a loading control 3 days after lentiviral infection. (e) MTH1 knock down impairs BJ-hTERT proliferation as compared to BJ fibroblasts. DIC images were captured 12 days following lentiviral infection with a 10x objective.

Supplementary Figure 6 Telomerase activity on terminal primers with 8-oxoG opposite rA.

(a) Telomerase reactions were conducted using primers containing three TTAGGG repeats terminating in GGG, GGGoxoG, or GGGToxoG (Supplemental Table 1, oligos #3, 9 and 10). Reactions contained cellular dNTP concentrations and 0.3 μM 32P-α-dGTP. Products were separated on denaturing gels. The LC was a 32P end labeled 36-mer oligonucleotide. Numbers with plus sign indicate number of added repeats. (b) and (c), Products were normalized to the LC, and used to calculate processivity (b) and relative activity (c). Bars represent the mean ± s.d. from three independent reactions. (d) Telomerase reactions contained high dNTPs (lane 1) or 2.9 μM dGTP (lanes 2-4) and 0.3 μM 32P-α-dGTP. Products were separated on polyacrylamide denaturing gels. The LC was 32P end-labeled 18-mer oligonucleotide. Bands representing addition (+) or subtraction (-) of telomeric repeats are indicated. Product size and corresponding sequence are shown, red = radiolabeled G insertion. (e) Products were quantitated and normalized to the LC, and used to calculate relative activity. Bars represent the mean and s.d. from three independent reactions.

Supplementary Figure 7 Telomerase-associated nuclease removes a primer terminal 8-oxoG.

(a) Telomerase preparations lack contaminating exonuclease activity. 8-oxoG substitutions in the 4R substrate are indicated in red “Go” (oligos 6-8 in Supplementary Table 1). Reactions (20 μl) contained 19.5 pmol primer and 0.5 pmol 32P end-labeled primer for a final total concentration of 1 μM in 1x human telomere buffer. The dNTPs were omitted. Reactions were initiated by adding 6 μl telomerase extract and incubated for 60 min at 30oC, and then run on polyacrylamide denaturing gels. (b) Telomerase reactions were conducted using primers containing three or four TTAGGG repeats, with a 3’ terminal G or 8-oxoG (oligos 3, 4, 6 and 7 Supplementary Table 1). Reactions contained high dNTPs (lane 1) or 2.9 μM dGTP (lanes 2-5) and 0.3 μM 32P-α-dGTP. Products were separated on polyacrylamide denaturing gels. The LC was 32P end-labeled 18-mer oligonucleotide. Bands representing addition (+) or subtraction (-) of telomeric repeats are indicated for 3 repeat (left) and 4 repeat (right) substrate. Product size and corresponding sequence are shown, red = radiolabeled G insertion. (c) Products were quantitated and normalized to the LC, and used to calculate relative activity. Bars represent the mean and s.d. from three independent reactions. Asterisks, ** p < 0.01, *** p < 0.001 base on two-tailed Student’s t test.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Table 1 (PDF 1796 kb)

Supplementary Data Set 1

Uncropped gels and blot images from main figures (PDF 22741 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fouquerel, E., Lormand, J., Bose, A. et al. Oxidative guanine base damage regulates human telomerase activity. Nat Struct Mol Biol 23, 1092–1100 (2016). https://doi.org/10.1038/nsmb.3319

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3319

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing