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:

Tet3 regulates synaptic transmission and homeostatic plasticity via DNA oxidation and repair

Abstract

Contrary to the long-held belief that DNA methylation of terminally differentiated cells is permanent and essentially immutable, post-mitotic neurons exhibit extensive DNA demethylation. The cellular function of active DNA demethylation in neurons, however, remains largely unknown. Tet family proteins oxidize 5-methylcytosine to initiate active DNA demethylation through the base-excision repair (BER) pathway. We found that synaptic activity bi-directionally regulates neuronal Tet3 expression. Functionally, knockdown of Tet or inhibition of BER in hippocampal neurons elevated excitatory glutamatergic synaptic transmission, whereas overexpressing Tet3 or Tet1 catalytic domain decreased it. Furthermore, dysregulation of Tet3 signaling prevented homeostatic synaptic plasticity. Mechanistically, Tet3 dictated neuronal surface GluR1 levels. RNA-seq analyses further revealed a pivotal role of Tet3 in regulating gene expression in response to global synaptic activity changes. Thus, Tet3 serves as a synaptic activity sensor to epigenetically regulate fundamental properties and meta-plasticity of neurons via active DNA demethylation.

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

Access options

Buy this article

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

Figure 1: Synaptic activity–dependent expression of Tet3 regulates glutamatergic synaptic transmission.
Figure 2: DNA oxidation and BER regulates glutamatergic synaptic transmission.
Figure 3: Tet3 signaling mediates homeostatic synaptic scaling-up of glutamatergic synaptic transmission.
Figure 4: Tet3 signaling mediates bicuculline-induced homeostatic synaptic scaling-down of glutamatergic synaptic transmission.
Figure 5: Tet3 signaling regulates neuronal surface GluR1 levels.
Figure 6: Tet3 regulates gene expression in neurons in response to global synaptic activity changes.
Figure 7: Essential role of Tet3 in neuronal activity–induced DNA methylation dynamics at the Bdnf IV promoter region and gene expression.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Tsankova, N., Renthal, W., Kumar, A. & Nestler, E.J. Epigenetic regulation in psychiatric disorders. Nat. Rev. Neurosci. 8, 355–367 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Gräff, J., Kim, D., Dobbin, M.M. & Tsai, L.H. Epigenetic regulation of gene expression in physiological and pathological brain processes. Physiol. Rev. 91, 603–649 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Telese, F., Gamliel, A., Skowronska-Krawczyk, D., Garcia-Bassets, I. & Rosenfeld, M.G. “Seq-ing” insights into the epigenetics of neuronal gene regulation. Neuron 77, 606–623 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shin, J., Ming, G.L. & Song, H. DNA modifications in the mammalian brain. Phil. Trans. R. Soc. Lond. B 369 (2014).

  5. Day, J.J., Kennedy, A.J. & Sweatt, J.D. DNA methylation and its implications and accessibility for neuropsychiatric therapeutics. Annu. Rev. Pharmacol. Toxicol. 55, 591–611 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Law, J.A. & Jacobsen, S.E. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 11, 204–220 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Martinowich, K. et al. DNA methylation-related chromatin remodeling in activity-dependent BDNF gene regulation. Science 302, 890–893 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Miller, C.A. & Sweatt, J.D. Covalent modification of DNA regulates memory formation. Neuron 53, 857–869 (2007).

    CAS  PubMed  Google Scholar 

  10. Nelson, E.D., Kavalali, E.T. & Monteggia, L.M. Activity-dependent suppression of miniature neurotransmission through the regulation of DNA methylation. J. Neurosci. 28, 395–406 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ma, D.K. et al. Neuronal activity-induced Gadd45b promotes epigenetic DNA demethylation and adult neurogenesis. Science 323, 1074–1077 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Feng, J. et al. Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat. Neurosci. 13, 423–430 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Guo, J.U., Su, Y., Zhong, C., Ming, G.L. & Song, H. Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain. Cell 145, 423–434 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guo, J.U. et al. Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat. Neurosci. 14, 1345–1351 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lister, R. et al. Global epigenomic reconfiguration during mammalian brain development. Science 341, 1237905 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Guo, J.U., Su, Y., Zhong, C., Ming, G.L. & Song, H. Emerging roles of TET proteins and 5-hydroxymethylcytosines in active DNA demethylation and beyond. Cell Cycle 10, 2662–2668 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guo, J.U. et al. Genome-wide antagonism between 5-hydroxymethylcytosine and DNA methylation in the adult mouse brain. Front. Biol. (Beijing) 9, 66–74 (2014).

    Article  CAS  Google Scholar 

  18. Tahiliani, M. et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ito, S. et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466, 1129–1133 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pollen, A.A. et al. Low-coverage single-cell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex. Nat. Biotechnol. 32, 1053–1058 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Pastor, W.A., Aravind, L. & Rao, A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat. Rev. Mol. Cell Biol. 14, 341–356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kriaucionis, S. & Heintz, N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324, 929–930 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Szulwach, K.E. et al. 5-hmC–mediated epigenetic dynamics during postnatal neurodevelopment and aging. Nat. Neurosci. 14, 1607–1616 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yao, B. et al. Genome-wide alteration of 5-hydroxymethylcytosine in a mouse model of fragile X-associated tremor/ataxia syndrome. Hum. Mol. Genet. 23, 1095–1107 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Kaas, G.A. et al. TET1 controls CNS 5-methylcytosine hydroxylation, active DNA demethylation, gene transcription and memory formation. Neuron 79, 1086–1093 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Rudenko, A. et al. Tet1 is critical for neuronal activity-regulated gene expression and memory extinction. Neuron 79, 1109–1122 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Todarello, G. et al. Incomplete penetrance of NRXN1 deletions in families with schizophrenia. Schizophr. Res. 155, 1–7 (2014).

    Article  PubMed  Google Scholar 

  28. Williams, K. et al. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 473, 343–348 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chen, Q., Chen, Y., Bian, C., Fujiki, R. & Yu, X. TET2 promotes histone O-GlcNAcylation during gene transcription. Nature 493, 561–564 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Ito, S. et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 333, 1300–1303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. He, Y.F. et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science 333, 1303–1307 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hajkova, P. et al. Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science 329, 78–82 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Turrigiano, G.G. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell 135, 422–435 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Aoto, J., Nam, C.I., Poon, M.M., Ting, P. & Chen, L. Synaptic signaling by all-trans retinoic acid in homeostatic synaptic plasticity. Neuron 60, 308–320 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lee, H.K. Ca-permeable AMPA receptors in homeostatic synaptic plasticity. Front. Mol. Neurosci. 5, 17 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Koike, M., Iino, M. & Ozawa, S. Blocking effect of 1-naphthyl acetyl spermine on Ca2+-permeable AMPA receptors in cultured rat hippocampal neurons. Neurosci. Res. 29, 27–36 (1997).

    Article  CAS  PubMed  Google Scholar 

  37. Shepherd, J.D. et al. Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron 52, 475–484 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gao, M. et al. A specific requirement of Arc/Arg3.1 for visual experience-induced homeostatic synaptic plasticity in mouse primary visual cortex. J. Neurosci. 30, 7168–7178 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rial Verde, E.M., Lee-Osbourne, J., Worley, P.F., Malinow, R. & Cline, H.T. Increased expression of the immediate-early gene arc/arg3.1 reduces AMPA receptor–mediated synaptic transmission. Neuron 52, 461–474 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Poo, M.M. Neurotrophins as synaptic modulators. Nat. Rev. Neurosci. 2, 24–32 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Rutherford, L.C., Nelson, S.B. & Turrigiano, G.G. BDNF has opposite effects on the quantal amplitude of pyramidal neuron and interneuron excitatory synapses. Neuron 21, 521–530 (1998).

    Article  CAS  PubMed  Google Scholar 

  42. Madabhushi, R., Pan, L. & Tsai, L.H. DNA damage and its links to neurodegeneration. Neuron 83, 266–282 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gu, T.P. et al. The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature 477, 606–610 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Shen, L. et al. Tet3 and DNA replication mediate demethylation of both the maternal and paternal genomes in mouse zygotes. Cell Stem Cell 15, 459–470 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Guo, F. et al. Active and passive demethylation of male and female pronuclear DNA in the Mammalian zygote. Cell Stem Cell 15, 447–458 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Xu, Y. et al. Tet3 CXXC domain and dioxygenase activity cooperatively regulate key genes for Xenopus eye and neural development. Cell 151, 1200–1213 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hahn, M.A. et al. Dynamics of 5-hydroxymethylcytosine and chromatin marks in Mammalian neurogenesis. Cell Reports 3, 291–300 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Zhang, R.R. et al. Tet1 regulates adult hippocampal neurogenesis and cognition. Cell Stem Cell 13, 237–245 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ibata, K., Sun, Q. & Turrigiano, G.G. Rapid synaptic scaling induced by changes in postsynaptic firing. Neuron 57, 819–826 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Abraham, W.C. & Bear, M.F. Metaplasticity: the plasticity of synaptic plasticity. Trends Neurosci. 19, 126–130 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Song, H., Stevens, C.F. & Gage, F.H. Astroglia induce neurogenesis from adult neural stem cells. Nature 417, 39–44 (2002).

    CAS  PubMed  Google Scholar 

  52. Ge, S. et al. GABA regulates synaptic integration of newly generated neurons in the adult brain. Nature 439, 589–593 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Turrigiano, G.G., Leslie, K.R., Desai, N.S., Rutherford, L.C. & Nelson, S.B. Activity-dependent scaling of quantal amplitude in neocortical neurons. Nature 391, 892–896 (1998).

    Article  CAS  PubMed  Google Scholar 

  54. Jang, M.H. et al. Secreted frizzled-related protein 3 regulates activity-dependent adult hippocampal neurogenesis. Cell Stem Cell 12, 215–223 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Robinson, M.D., McCarthy, D.J. & Smyth, G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Song, H.J., Stevens, C.F. & Gage, F.H. Neural stem cells from adult hippocampus develop essential properties of functional CNS neurons. Nat. Neurosci. 5, 438–445 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Kim, J.Y. et al. DISC1 regulates new neuron development in the adult brain via modulation of AKT-mTOR signaling through KIAA1212. Neuron 63, 761–773 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. Huganir, P. Worley, G.C. Turrigiano and L. Chen for suggestions, members of the Ming and Song laboratories for help and critical comments, G.L. Xu for Tet3 antibodies, and L. Liu, Q. Hussaini and Y. Cai for technical support. This work was supported by the US National Institutes of Health (NS047344 to H.S., NS048271 and MH105128 to G.-l.M., NS062691 to G.C. and D.H.G.), a grant from the Simons Foundation (SFARI240011 to H.S.), the Brain and Behavior Research Foundation (H.S., G.-l.M. and Y.S.), Maryland Stem Cell Research Foundation (G.-l.M. and H.S.), and by the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (G.-l.M., D.H.G. and G.C.). J.S. was supported by a Samsung fellowship.

Author information

Authors and Affiliations

Authors

Contributions

H.Y. performed electrophysiological analyses. Y.S. performed biochemical and DNA methylation analyses. J.S. and J.U.G. performed bioinformatics analysis. C.Z. and Y.-L.W. generated AAV. F.G., D.H.G. and G.C. performed RNA-seq. G.-l.M. and H.S. designed the project and wrote the manuscript.

Corresponding author

Correspondence to Hongjun Song.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Basic characterization of neurons with Tet3 knockdown, overexpression of Tet1-CD or Tet1-mCD.

(a) Nuclear localization of Tet3 protein. Shown is a sample image of immunostaining of hippocampal neurons for Tet3, synapsin I and DAPI. Scale bar: 10 μm. (b) Schematic diagrams of AAV constructs used to infect cultured hippocampal neurons. (c) Sample images of neuronal cultures infected with AAV co-expressing EYFP and shRNAs. Scale bar: 20 μm. Note that over 99% neurons were EYFP+, indicating very high infection efficacy. Also shown is q-PCR analysis of mRNA expression of Tet1, 2, 3 after AAV-mediated expression of different shRNAs. Value represent mean + s.e.m. (n = 3; *P < 0.01; ANOVA; P = 0.03, sh-control vs sh-Tet1; P = 0.002, sh-control vs sh-Tet2; P = 0.01, sh-control vs sh-Tet3-1; P = 0.01 sh-control vs sh-Tet3-2). (d) Dot blot analysis of 5hmC levels in neurons under different conditions. Different cultures were treated with TTX (1 μM) or bicuculline (20 μM) for 48 hours before analyses. (e) Lack of differences in the excitability of neurons with or without Tet3 KD. Shown are sample current-clamp whole-cell recording traces in response to injection of different amount of currents (top panels) and summary of numbers of action potentials (bottom panel). Values represent mean + s.e.m. (f) Lack of differences in the density of synapsin 1+ boutons in neurons upon Tet3 knockdown or Tet1-CD expression. Shown on the top panel are sample confocal images of Tuj1 and synapsin I immunostaining and DAPI staining. Scale bar: 50 μm. Shown on the bottom panel is a summary of density of synapsin I+ boutons on Tuj1+ neurites. Values represent mean + s.e.m. (n > 3 cultures).

Supplementary Figure 2 Rank plots of mEPSC amplitudes to calculate scaling factors for neurons with Tet3 KD under different conditions.

(a) Tet3 KD scaled up mEPSC amplitudes multiplicatively. Shown are plots of ranked mEPSC amplitudes recorded from neurons expressing sh-Tet3-1 (left) or sh-Tet3-2 (right) against ranked mEPSC amplitudes from neurons expressing sh-control. The data are best described by a multiplicative increase in mEPSC amplitude with a scaling factor of 1.48 (sh-Tet3-1) and 1.98 (sh-Tet3-2). (b-e) Lack of significant TTX-induced scaling-up (b-c) and bicuculline-induced scaling-down (d-e) in Tet3 KD neurons. Similar as in (a), shown are plots of ranked mEPSC amplitudes recorded from neurons expressing different shRNAs with TTX (1 μM; b-c) or bicuculline treatment (20 μM; d-e) for 48 hours against ranked mEPSC amplitudes from neurons with the saline treatment. Scaling factor for each condition is listed at the top left corner. (f-g) RA (1 μm, 2 hour treatment) and Tet3 KD induced scaling-up occludes each other.

Supplementary Figure 3 Rank plots of mEPSC amplitudes to calculate scaling factors for neurons with treatment of BER inhibitors.

(a) Plots of ranked mEPSC amplitudes from neurons with treatment of ABT (50 μM, left) or CRT (50 μM, right) for 48 hours against ranked mEPSC amplitudes from neurons with the vehicle treatment. The data are best described by a multiplicative increase in mEPSC amplitude with a scaling factor of 1.40 (ABT) and 1.45 (CRT). (b-e) Lack of significant TTX-induced scaling up (b-c) and bicuculline-induced scaling-down (d-e) upon inhibition of DNA repair. Similar as in (a), shown are plots of ranked mEPSC amplitudes recorded from neurons under different drug treatments that were concurrently treated with TTX (1 μM; b-c) or bicuculline (20 μM; d-e) for 48 hours against ranked mEPSC amplitudes from those neurons without the TTX or bicuculline treatment. Scaling factor for each condition is listed at the top left corner.

Supplementary Figure 4 Rank plots of mEPSC amplitudes to calculate scaling factors for neurons with overexpression of Tet3.

(a) Plot of ranked mEPSC amplitudes from neurons co-expressing EYFP and Tet3 against ranked mEPSC amplitudes from neurons expressing EYFP alone. The data are best described by a multiplicative decrease in mEPSC amplitude with a scaling factor of 0.47. (b-e) Lack of significant TTX-induced scaling up (b-c) and bicuculline-induced scaling-down (d-e) upon Tet3 overexpression. Similar as in (a), shown are plots of ranked mEPSC amplitudes recorded from neurons over-expressing Tet3 (b, d) or EYFP (c, e) with TTX (1 μM; b-c) or bicuculline treatment (20 μM; d-e) for 48 hours against ranked mEPSC amplitudes from those neurons with the saline treatment. Scaling factor for each condition is listed at the top left corner.

Supplementary Figure 5 Rank plots of mEPSC amplitudes to calculate scaling factors for neurons with overexpression of Tet1-CD or Tet1-mCD.

(a) Plot of ranked mEPSC amplitudes from neurons co-expressing EYFP and Tet1-CD against ranked mEPSC amplitudes from neurons expressing EYFP alone with a scaling factor of 0.77. (b-e) Lack of significant TTX-induced scaling up (b-c) and bicuculline-induced scaling-down (d-e) upon Tet1-CD expression. Similar as in (a), shown are plots of ranked mEPSC amplitudes recorded from neurons over-expressing Tet1-CD (b, d) or EYFP (c, e) with TTX (1 μM; b-c) or bicuculline treatment (20 μM; d-e) for 48 hours against ranked mEPSC amplitudes from those neurons with the saline treatment. Scaling factor for each condition is listed at the top left corner.

Supplementary Figure 6 Total GluR1 and Arc protein levels under different conditions.

Shown are sample cropped Western blot images and quantification of total GluR1 (a) or Arc (b) levels under different conditions. The same biological samples as in Fig. 5c were used for a. Values represent mean + s.e.m. (n = 3 experiments; *P < 0.05; #P > 0.1; ANOVA; b: P = 0.002, sh-control vs sh-control + TTX; P = 0.007, sh-control vs sh-control + Bicu; P = 0.03, sh-control vs sh-Tet3-2). Full-length blots are presented in Supplementary Figure 11.

Supplementary Figure 7 Functional change of GluR2-lacking AMPA receptors upon changes in Tet3 expression and DNA demethylation signalling pathway.

(a) Lack of changes of surface or total GluR2 levels upon Tet3 KD or Tet1-CD expression. Shown are sample cropped Western blot images and quantification of surface and total GluR2 levels under different conditions. The same biological samples were used. Values represent mean + s.e.m. (n = 3). Full-length blots are presented in Supplementary Figure 11. (b) Effect of NASPM treatment on the mEPSC amplitude of neurons expressing sh-control or sh-Tet3-2. Values represent mean + s.e.m. (*P < 0.05; **P < 0.01; ANOVA; P = 0.03, before vs after in sh-control group; P = 0.003, before vs after in sh-Tet3-2 group). (c) Summary of decay time of mEPSCs recorded under different conditions. Same cells as in Fig. 1c-d & 2b-c. Values represent mean + s.e.m. (***P < 0.001; **P < 0.01; *P < 0.05; ANOVA; Left: P = 0.000006, sh-control vs sh-Tet3-1; P = 0.00005, sh-control vs sh-Tet3-2; Middle: P = 0.001, EYFP vs EYFP/Tet3 OE; P = 0.03, EYFP vs Tet1-CD; P = 0.02, Tet1-CD vs Tet1-mCD; right: P = 0.02, vehicle vs ABT; P = 0.002, vehicle vs CRT).

Supplementary Figure 8 RNA-seq analysis of neurons under different conditions.

(a) Confirmation of Tet3 knockdown efficacy upon AAV-mediated expression of sh-Tet3-2 in samples for RNA-seq. FPKM reads from three samples each are shown. (b) Heat-map of Pearson correlations among different RNA-seq samples. A total of 17 samples were subject to RNA-seq analysis and the top 2000 highly expressed genes were used for calculation of Pearson correlation. (c) Multidimensional scaling plot. edgeR was used for the analysis of 17 RNA-seq samples. Note clustering of biological replicates, but clear segregation of gene expression from neurons expressing sh-control and neurons expressing sh-Tet3-2. For neurons expressing sh-control, TTX and bicuculline treatment induced the opposite direction of gene expression alterations. Importantly, in Tet3-KD neurons, such changes were either attenuated (for bicuculline treatment) or abolished (for TTX treatment).

Supplementary Figure 9 Tet3 regulates expression of genes associated with synapses and synaptic transmission.

Pathway diagrams are modified from Kyoto Encyclopedia of Genes and Genomes (KEGG). Genes that exhibited significant changes of expression in Tet3-KD neurons (FDR < 0.05) are indicated by stars (red: genes activated by Tet3; blue: genes inhibited by Tet3). Data is based on RNA-seq analyses of neurons expressing sh-Tet3-2 compared to those expressing sh-control (n = 3 samples each).

Supplementary Figure 10 Lack of effect of Tet3 KD on immediate early genes.

(a) Q-PCR analysis of expression of immediate early genes in neurons expressing sh-control or sh-Tet3-2 at 4 hours after bicuculline treatment (20 μM). Values represent mean + s.e.m. (n = 3; *** P < 0.001; **P < 0.01; *P < 0.05; #P > 0.1; Student’s t-test; Arc: P = 0.02, vehicle vs Bicu in sh-control group; P = 0.01, vehicle vs Bicu in sh-Tet3-2 group; Egr4: P = 0.02, vehicle vs Bicu in sh-control group; P = 0.03, vehicle vs Bicu in sh-Tet3-2 group; c-fos: P = 0.006, vehicle vs Bicu in sh-control group; P = 0.00001, vehicle vs Bicu in sh-Tet3-2 group; Npas4: P = 0.002, vehicle vs Bicu in sh-control group; P = 0.02, vehicle vs Bicu in sh-Tet3-2 group). (b) Methylation status of Arc and Npas4 promoters and intragenic regions in neurons expressing sh-control or sh-Tet3-2. Each row represents one allele showing methylation status of individual CpG sites (open circle: unmethylated; closed circle methylated).

Supplementary Figure 11 Full length blots sample blots shown in other figures.

Shown are full blots for cropped sample gel images shown in Figures 1b, 5c, 7c and Supplementary Figures 6a, 6b and 7a.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 (PDF 1424 kb)

Supplementary Methods Checklist (PDF 387 kb)

Supplementary Table 1

Summary of shRNA and primer sequences used in the current study. (a) Primer sequences used for qPCR analysis; (b) shRNA sequences; (c) Primer sequences for bisulfite sequencing analysis; (d) Primer sequences for ChIP-PCR analyses. (XLS 41 kb)

Supplementary Table 2

Summary of RNA-seq analysis. (a) RNA-seq run and mapping information; (b) Gene list and information on differentially expressed genes upon Tet3 knockdown. (c-d) Gene lists and information on bicuculline-regulated genes in neurons expressing sh-control (c) and those expressing sh-Tet3-2 (d). (e-f) Gene lists and information on TTX-regulated genes in neurons expressing shRNA-control (e) and those expressing sh-Tet3-2 (f). (g) KEGG pathway analysis. (XLS 904 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, H., Su, Y., Shin, J. et al. Tet3 regulates synaptic transmission and homeostatic plasticity via DNA oxidation and repair. Nat Neurosci 18, 836–843 (2015). https://doi.org/10.1038/nn.4008

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4008

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