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:

Regulation of axon regeneration by the RNA repair and splicing pathway

Abstract

Mechanisms governing a neuron's regenerative ability are important but not well understood. We identify Rtca (RNA 3′-terminal phosphate cyclase) as an inhibitor of axon regeneration. Removal of Rtca cell-autonomously enhanced axon regrowth in the Drosophila CNS, whereas its overexpression reduced axon regeneration in the periphery. Rtca along with the RNA ligase Rtcb and its catalyst Archease operate in the RNA repair and splicing pathway important for stress-induced mRNA splicing, including that of Xbp1, a cellular stress sensor. Drosophila Rtca and Archease had opposing effects on Xbp1 splicing, and deficiency of Archease or Xbp1 impeded axon regeneration in Drosophila. Moreover, overexpressing mammalian Rtca in cultured rodent neurons reduced axonal complexity in vitro, whereas reducing its function promoted retinal ganglion cell axon regeneration after optic nerve crush in mice. Our study thus links axon regeneration to cellular stress and RNA metabolism, revealing new potential therapeutic targets for treating nervous system trauma.

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: Drosophila Rtca loss of function enhances axon regeneration in the CNS and PNS.
Figure 2: Drosophila Rtca overexpression reduces axon regeneration in the PNS.
Figure 3: Drosophila Rtca overexpression reduces motor axon regeneration.
Figure 4: The expression pattern of Rtca in Drosophila.
Figure 5: The interaction of Drosophila Rtca with known regeneration regulators.
Figure 6: The Drosophila Rtca pathway in regulating axon regeneration.
Figure 7: Rtca is an inhibitory factor for axon growth in vitro and RGC regeneration in vivo in rodents.

Similar content being viewed by others

References

  1. Filbin, M.T. Recapitulate development to promote axonal regeneration: good or bad approach? Phil. Trans. R. Soc. Lond. B 361, 1565–1574 (2006).

    Article  CAS  Google Scholar 

  2. Fitch, M.T. & Silver, J. CNS injury, glial scars, and inflammation: Inhibitory extracellular matrices and regeneration failure. Exp. Neurol. 209, 294–301 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Goldberg, J.L., Klassen, M.P., Hua, Y. & Barres, B.A. Amacrine-signaled loss of intrinsic axon growth ability by retinal ganglion cells. Science 296, 1860–1864 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Schwab, M.E. & Bartholdi, D. Degeneration and regeneration of axons in the lesioned spinal cord. Physiol. Rev. 76, 319–370 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Case, L.C. & Tessier-Lavigne, M. Regeneration of the adult central nervous system. Curr. Biol. 15, R749–R753 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Harel, N.Y. & Strittmatter, S.M. Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury? Nat. Rev. Neurosci. 7, 603–616 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Moore, D.L. et al. KLF family members regulate intrinsic axon regeneration ability. Science 326, 298–301 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Park, K.K. et al. Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science 322, 963–966 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Smith, P.D. et al. SOCS3 deletion promotes optic nerve regeneration in vivo. Neuron 64, 617–623 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sun, F. & He, Z. Neuronal intrinsic barriers for axon regeneration in the adult CNS. Curr. Opin. Neurobiol. 20, 510–518 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yiu, G. & He, Z. Glial inhibition of CNS axon regeneration. Nat. Rev. Neurosci. 7, 617–627 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chen, L. et al. Axon regeneration pathways identified by systematic genetic screening in C. elegans. Neuron 71, 1043–1057 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nix, P. et al. Axon regeneration genes identified by RNAi screening in C. elegans. J. Neurosci. 34, 629–645 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Song, Y. et al. Regeneration of Drosophila sensory neuron axons and dendrites is regulated by the Akt pathway involving Pten and microRNA bantam. Genes Dev. 26, 1612–1625 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bonilla, I.E., Tanabe, K. & Strittmatter, S.M. Small proline-rich repeat protein 1A is expressed by axotomized neurons and promotes axonal outgrowth. J. Neurosci. 22, 1303–1315 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Costigan, M. et al. Replicate high-density rat genome oligonucleotide microarrays reveal hundreds of regulated genes in the dorsal root ganglion after peripheral nerve injury. BMC Neurosci. 3, 16 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Fischer, D., Petkova, V., Thanos, S. & Benowitz, L.I. Switching mature retinal ganglion cells to a robust growth state in vivo: gene expression and synergy with RhoA inactivation. J. Neurosci. 24, 8726–8740 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nilsson, A., Moller, K., Dahlin, L., Lundborg, G. & Kanje, M. Early changes in gene expression in the dorsal root ganglia after transection of the sciatic nerve; effects of amphiregulin and PAI-1 on regeneration. Brain Res. Mol. Brain Res. 136, 65–74 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Veldman, M.B., Bemben, M.A., Thompson, R.C. & Goldman, D. Gene expression analysis of zebrafish retinal ganglion cells during optic nerve regeneration identifies KLF6a and KLF7a as important regulators of axon regeneration. Dev. Biol. 312, 596–612 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Araki, T. & Milbrandt, J. Ninjurin, a novel adhesion molecule, is induced by nerve injury and promotes axonal growth. Neuron 17, 353–361 (1996).

    Article  CAS  PubMed  Google Scholar 

  21. Genschik, P., Billy, E., Swianiewicz, M. & Filipowicz, W. The human RNA 3′-terminal phosphate cyclase is a member of a new family of proteins conserved in Eucarya, Bacteria and Archaea. EMBO J. 16, 2955–2967 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liu, K. et al. PTEN deletion enhances the regenerative ability of adult corticospinal neurons. Nat. Neurosci. 13, 1075–1081 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gabel, C.V., Antoine, F., Chuang, C.F., Samuel, A.D. & Chang, C. Distinct cellular and molecular mechanisms mediate initial axon development and adult-stage axon regeneration in C. elegans. Development 135, 1129–1136 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Popow, J. et al. HSPC117 is the essential subunit of a human tRNA splicing ligase complex. Science 331, 760–764 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Remus, B.S. & Shuman, S. A kinetic framework for tRNA ligase and enforcement of a 2′-phosphate requirement for ligation highlights the design logic of an RNA repair machine. RNA 19, 659–669 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Englert, M., Sheppard, K., Aslanian, A., Yates, J.R. III & Soll, D. Archaeal 3′-phosphate RNA splicing ligase characterization identifies the missing component in tRNA maturation. Proc. Natl. Acad. Sci. USA 108, 1290–1295 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Tanaka, N. & Shuman, S. RtcB is the RNA ligase component of an Escherichia coli RNA repair operon. J. Biol. Chem. 286, 7727–7731 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tanaka, N., Meineke, B. & Shuman, S. RtcB, a novel RNA ligase, can catalyze tRNA splicing and HAC1 mRNA splicing in vivo. J. Biol. Chem. 286, 30253–30257 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Desai, K.K., Cheng, C.L., Bingman, C.A., Phillips, G.N. Jr. & Raines, R.T. A tRNA splicing operon: Archease endows RtcB with dual GTP/ATP cofactor specificity and accelerates RNA ligation. Nucleic Acids Res. 42, 3931–3942 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Popow, J., Jurkin, J., Schleiffer, A. & Martinez, J. Analysis of orthologous groups reveals archease and DDX1 as tRNA splicing factors. Nature 511, 104–107 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Canaves, J.M. Predicted role for the archease protein family based on structural and sequence analysis of TM1083 and MTH1598, two proteins structurally characterized through structural genomics efforts. Proteins 56, 19–27 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Das, U. & Shuman, S. 2′-Phosphate cyclase activity of RtcA: a potential rationale for the operon organization of RtcA with an RNA repair ligase RtcB in Escherichia coli and other bacterial taxa. RNA 19, 1355–1362 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tanaka, N., Chakravarty, A.K., Maughan, B. & Shuman, S. Novel mechanism of RNA repair by RtcB via sequential 2′,3′-cyclic phosphodiesterase and 3′-phosphate/5′-hydroxyl ligation reactions. J. Biol. Chem. 286, 43134–43143 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chakravarty, A.K. & Shuman, S. The sequential 2′,3′-cyclic phosphodiesterase and 3′-phosphate/5′-OH ligation steps of the RtcB RNA splicing pathway are GTP-dependent. Nucleic Acids Res. 40, 8558–8567 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 8, 519–529 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. & Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881–891 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Jurkin, J. et al. The mammalian tRNA ligase complex mediates splicing of XBP1 mRNA and controls antibody secretion in plasma cells. EMBO J. 33, 2922–2936 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ryoo, H.D., Domingos, P.M., Kang, M.J. & Steller, H. Unfolded protein response in a Drosophila model for retinal degeneration. EMBO J. 26, 242–252 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Ramon y Cajal, S. Degeneration and Regeneration of the Nervous System (Oxford University Press, London, 1928).

  40. Liu, X., Hawkes, E., Ishimaru, T., Tran, T. & Sretavan, D.W. EphB3: an endogenous mediator of adult axonal plasticity and regrowth after CNS injury. J. Neurosci. 26, 3087–3101 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lu, Y., Belin, S. & He, Z. Signaling regulations of neuronal regenerative ability. Curr. Opin. Neurobiol. 27, 135–142 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hu, Y. et al. Differential effects of unfolded protein response pathways on axon injury-induced death of retinal ganglion cells. Neuron 73, 445–452 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Han, C., Jan, L.Y. & Jan, Y.N. Enhancer-driven membrane markers for analysis of nonautonomous mechanisms reveal neuron-glia interactions in Drosophila. Proc. Natl. Acad. Sci. USA 108, 9673–9678 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Grueber, W.B., Jan, L.Y. & Jan, Y.N. Different levels of the homeodomain protein cut regulate distinct dendrite branching patterns of Drosophila multidendritic neurons. Cell 112, 805–818 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Sepp, K.J., Schulte, J. & Auld, V.J. Peripheral glia direct axon guidance across the CNS/PNS transition zone. Dev. Biol. 238, 47–63 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Xiang, Y. et al. Light-avoidance-mediating photoreceptors tile the Drosophila larval body wall. Nature 468, 921–926 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Awasaki, T., Lai, S.L., Ito, K. & Lee, T. Organization and postembryonic development of glial cells in the adult central brain of Drosophila. J. Neurosci. 28, 13742–13753 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Song, W., Onishi, M., Jan, L.Y. & Jan, Y.N. Peripheral multidendritic sensory neurons are necessary for rhythmic locomotion behavior in Drosophila larvae. Proc. Natl. Acad. Sci. USA 104, 5199–5204 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Grueber, W.B. et al. Projections of Drosophila multidendritic neurons in the central nervous system: links with peripheral dendrite morphology. Development 134, 55–64 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Huang, H. et al. PTEN affects cell size, cell proliferation and apoptosis during Drosophila eye development. Development 126, 5365–5372 (1999).

    CAS  PubMed  Google Scholar 

  51. Ritzenthaler, S., Suzuki, E. & Chiba, A. Postsynaptic filopodia in muscle cells interact with innervating motoneuron axons. Nat. Neurosci. 3, 1012–1017 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Emoto, K., Parrish, J.Z., Jan, L.Y. & Jan, Y.N. The tumour suppressor Hippo acts with the NDR kinases in dendritic tiling and maintenance. Nature 443, 210–213 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Parrish, J.Z., Emoto, K., Kim, M.D. & Jan, Y.N. Mechanisms that regulate establishment, maintenance, and remodeling of dendritic fields. Annu. Rev. Neurosci. 30, 399–423 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Xiong, X. et al. Protein turnover of the Wallenda/DLK kinase regulates a retrograde response to axonal injury. J. Cell Biol. 191, 211–223 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ultanir, S.K. et al. Chemical genetic identification of NDR1/2 kinase substrates AAK1 and Rabin8 Uncovers their roles in dendrite arborization and spine development. Neuron 73, 1127–1142 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Younger, S.K. Ultanir, S. Yadav, D. Wang, S. Barbel, M. Tynan-La Fontaine and J. Sacramento for technical assistance; J. Martinez for advice on the Rtca pathway; I. Hariharan (University of California, Berkeley), T. Xu (Yale) and H.D. Ryoo (New York University) for fly lines; the Bloomington Stock Center, Kyoto Drosophila Genetic Resource Center (DGRC), Vienna Drosophila Resource Center (VDRC) and Exelixis for fly stocks; DGRC for plasmids; UCSF ES Cell Targeting Core for generating the knockout mouse; and members of the Jan laboratory for discussions. Y.S. is a recipient of a National Institute of Neurological Disorders and Stroke (NINDS) Pathway to Independence Award. This work was supported by a NINDS K99/R00 award (1K99NS088211-01) to Y.S., a National Science Foundation Graduate Research Fellowship (1144247) to S.M., a UC Irvine-Roman Reed Spinal Cord Injury grant (P0045665) to Y.N.J., a NIH grant (2R37NS040929) to Y.N.J., a NIH grant (EY010688-19) to D.S. and a Research to Prevent Blindness grant to D.S. L.Y.J. and Y.N.J. are investigators of Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

Y.S. carried out most of the experiments and performed the data analysis. D.S. performed the optic nerve crush experiment; E.A.S. performed β-galactosidase staining; J.B. performed Rtca expression analysis; X.H. contributed to the neuronal culture experiment; T.C. contributed to the RGC axon regeneration analysis; X.X. performed the motor axon regeneration assay; S.M. contributed to the stress assay; C.H. made the UAS-Rtca fly strain; T.-T.N. contributed to the optic nerve crush experiment; J.C.B. and M.S.B. contributed to the mouse axon regeneration experiment; and Y.S., L.Y.J. and Y.N.J. together conceived the research and wrote the manuscript.

Corresponding author

Correspondence to Yuh Nung Jan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 A schematic diagram depicting the axotomy protocol of da neuron axons in the PNS and CNS.

The axons in the PNS and CNS are labeled. Dashed line indicates VNC boundary. The axon terminal within the VNC of one representative class IV da neuron is shown in green.

Supplementary Figure 2 Drosophila Rtca loss of function, as in RtcaNP5057, which alters transcript expression, enhances axon regeneration in the VNC without affecting axon terminal patterning.

(a) P element insertion in dRtcaNP5057 disrupts mRNA splicing and reduces transcript level. Arrows indicate the primer locations. Two pairs of primers were used and are color coded in green and turquoise. The PCR products are color coded in the same way according to the primer position. Blots Gels were cropped and full-length images are presented in Supplementary Figure 13. (b) The overall patterning of class IV da neuron axons in the VNC (labeled by ppk-CD4tdGFP) is not affected after dRtca LOF. (c) Single clone analysis shows that dRtca LOF does not affect gross patterning of the nerve terminals. The axon terminal of a single class IV da neuron is shown in whole animal dRtca LOF mutants (upper panels), or in animals with dRtca RNAi in all class IV da neurons (lower panels). Scale bar = 20 μm.

Supplementary Figure 3 Quantification of sensory axon regeneration.

(a) For axon regeneration in the VNC, “Terminal branching” counts the number of axons that regenerate, reach the commissure bundle and appear to form elaborate branches, shown in red. “Commissure regrowth” counts the number of axons that have regenerated to connect the boundaries of commissure segments, longitudinally or laterally, shown in blue. (b) For axon regeneration in the PNS, “Regeneration index” is calculated as an increase of “axon length”/“distance between the cell body and the axon converging point (DCAC)”.

Supplementary Figure 4 Analyses of the CNS regeneration phenotype in Drosophila Rtca and Pten pathway mutants, as well as PNS axon regeneration after Rac1DN overexpression.

(a, b) Double mutants of dRtca and Pten do not further promote axon regeneration in the VNC. (a) Regeneration percentage is not significantly different between dRtca single mutants and dRtca Pten double mutants (N = 46, 16, 12 lesioned segments; ***P < 0.001, Fisher’s exact test, P < 0.0001, P = 0.0005, P = 0.1746). (b) Terminal branching and Commissure regrowth are comparable between dRtca single mutants and dRtca Pten double mutants (N = 28, 8, 6 larvae; *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA followed by Holm-Sidak's multiple comparisons test and Tukey's multiple comparisons test, respectively). (c, d) The regeneration phenotype in dRtca LOF mutants appears to be comparable if not stronger than Pten mutants (PtenMGH6, a hypomorphic allele) or Akt overexpression (ppk-Gal4>Akt) (N = 46, 17, 13, 16, 14 lesioned segments; **P < 0.01, ***P < 0.001, Fisher’s exact test in c, P = 0.0027, P = 0.0002, P = 0.0001, P < 0.0001; N = 29, 9, 7, 8, 7 larvae; *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA followed by Holm-Sidak's multiple comparisons test and Dunn’s multiple comparisons test, respectively, in d). (eg) Rac1DN overexpression in class IV da neurons does not affect axon regeneration in the periphery (N = 27, 20 neurons; P > 0.05, Fisher’s exact test in e, P = 0.7365, and two-tailed unpaired Student’s t-test in f and g, P = 0.3533, P = 0.6708). Data are expressed as mean ± s.e.m. in bar plots or as median with minimum and maximum values in box-and-whisker plots, and box limits represent interquartile range.

Supplementary Figure 5 The proposed model of the Rtca-Archease-Xbp1 pathway in the regulation of regeneration.

Axon damage results in some type of cellular stress and the repair/splicing of mRNA substrates, such as Xbp1. The 3′-phosphate of the spliced RNA is converted into 2′,3′-cyclic phosphate by Rtca, whereas the RNA ligase Rtcb reverses this reaction and re-ligates the processed RNA, with the help of its catalyst Archease. Thus Rtca and Archease may have opposing functions on RNA repair/splicing and on regeneration.

Supplementary Figure 6 Drosophila ArcheasePBc01013 leads to altered mRNA splicing and reduced transcript expression, and is thus a loss of function allele.

Arrows indicate the primer locations. Two pairs of primers were used and are color coded in green and turquoise. The PCR products are color coded in the same way according to the primer position. Blots Gels were cropped and full-length images are presented in Supplementary Figure 14.

Supplementary Figure 7 Xbp1s overexpression increases class III da neuron axon regeneration in the periphery.

Dashed circle marks the injury site and arrowheads show the regenerating axon (N = 19, 24 neurons; *P < 0.05, Fisher’s exact test, P = 0.0261 and two-tailed unpaired Student’s t-test, P = 0.0384). Scale bar = 20 μm. Data are expressed as median with minimum and maximum values in box-and-whisker plots, and box limits represent interquartile range.

Supplementary Figure 8 The Rtca mutant mouse.

(a) A knockout-first line with a LacZ cassette inserted into the Rtca locus was used to generate the Rtca mutant mouse. The resulting mouse RtcaLacZ_loxP (RtcaIns/Ins) has a lacZ cassette inserted in-between the 3rd and 4th exons of Rtca, meanwhile, the 4th exon is flanked by loxP sites. (b) The lacZ insertion disrupts Rtca splicing and dramatically reduces transcript level, and thus results in a hypomorphic allele. Arrows indicate the primer locations. Two pairs of primers were used and are color coded in green and turquoise. The PCR products are color coded in the same way according to the primer position. Blots Gels were cropped and full-length images are presented in Supplementary Figure 15. Quantification of Rtca mRNA shows about 81% reduction (N = 4 experiments; the expression level of each data point was normalized to the average value of WT; **P < 0.01, two-tailed unpaired Student’s t-test, P = 0.0035). (c) Western blot analysis using retina tissue was performed. Anti-Rtca HPA027982 recognizes amino acids 111-193. The antibody cleanly labels the Rtca protein in WT, which is 39 KD, and shows a dramatic reduction of the protein after the insertion site in RtcaIns/Ins mutants, confirming the reduction of the Rtca full-length protein. The green line indicates the antigen region. Quantification of Rtca protein shows about 82% reduction (N = 3 experiments; the expression level of each data point was normalized to the average value of WT; *P < 0.05, two-tailed unpaired Student’s t-test, P = 0.0252). Data are expressed as mean ± s.e.m.

Supplementary Figure 9 Rtca LOF during development does not affect adult RGC number or survival after injury.

(a–c) Three weeks after optic nerve crush of sibling controls (Rtca+/+ and RtcaIns/+) and RtcaIns/Ins mutant mice, retina sections from both the control eye (uninjured nerve) and injured eye (crushed nerve) were immunostained for Tuj1 (βIII tubulin) to label RGC and DAPI to label nuclei. Confocal imaging of retina sections from mutant animals, normalized to siblings, shows no differences in basal RGC number in the contralateral uninjured retinas in (a, N = 3 siblings and 3 RtcaIns/Ins mice) or in RGC survival three weeks after optic nerve crush (b, N = 4 siblings and 5 RtcaIns/Ins mice; P > 0.05, two-tailed unpaired Student’s t-test, P = 0.6776, P = 0.9249). Representative confocal images of the retina sections immunostained for Tuj1 and DAPI are shown in (c). Scale bar = 20 μm. Data are expressed as mean ± s.e.m.

Supplementary Figure 10 Rtca is expressed in retinal ganglion cells.

(a–f) Positive LacZ staining was observed in the RGC layer of RtcaIns/+ mice (ac, arrow) but not in WT littermates (df). (g) β-Galactosidase staining in RtcaIns/+ mice shows distinct expression of the LacZ reporter within NeuN+ neurons in the retinal ganglion cell layer of the retina, which is absent in WT littermates, indicating that Rtca is expressed by RGCs. Scale bar = 20 μm.

Supplementary Figure 11 Rtca reduction enhances RGC axon regeneration after optic nerve crush.

(ad) Three weeks after optic nerve crush in 8–12 weeks old adult siblings (Rtca+/+ and RtcaIns/+), and RtcaIns/Ins mutant mice, regenerating fibers were labeled by anti-NF200 immunostaining. (a, b) Tissue sections through the optic nerve of a RtcaIns/Ins mutant mice show regenerating axons up to 300 μm distal to the lesion site, whereas no obvious axon regrowth beyond the crush site is seen in similar sections from a sibling control (RtcaIns/+, asterisk marks the crush site). Boxed regions are shown at higher magnification. Curving, turning and looping of axons were observed, indicative of new axon growth (arrowheads). (c) The furthest distance that axons travelled beyond the injury site is about 3.5 times longer in the mutants compared to siblings. (d) Regenerating fibers were counted at specified distances from the lesion site. More fibers regenerate in RtcaIns/Ins mice compared to sibling controls (N = 7 siblings and 4 RtcaIns/Ins mice; ***P < 0.001, two-tailed unpaired Student’s t-test in c; **P < 0.01, two-way ANOVA in d, P = 0.0012). (e) The optic nerve crush site is marked by the presence of ED1 staining, which labels the infiltrating macrophages. Whereas axons (labeled by NF200) rarely grow through the ED1+ region in the sibling controls (RtcaIns/+), a large number of axons are seen hundreds of microns beyond the ED1+ region in RtcaIns/Ins mutants. Asterisks mark the crush site. Scale bar = 100 μm. Data are expressed as mean ± s.e.m. in bar plots or as median with minimum and maximum values in box-and-whisker plots, and box limits represent interquartile range.

Supplementary Figure 12 Full-length DNA agarose gel for the analysis of Xbp1 splicing.

Full length DNA agarose gel (corresponding to Figure 6g).

Supplementary Figure 13 Full-length DNA agarose gel for the analysis of Drosohila Rtca expression.

Full length DNA agarose gel (corresponding to Supplementary Figure 2a).

Supplementary Figure 14 Full-length DNA agarose gel for the analysis of Drosophila Archease expression.

Full length DNA agarose gel (corresponding to Supplementary Figure 6).

Supplementary Figure 15 Full-length DNA agarose gel for the analysis of mouse Rtca expression.

Full length DNA agarose gel (corresponding to Supplementary Figure 8b).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Song, Y., Sretavan, D., Salegio, E. et al. Regulation of axon regeneration by the RNA repair and splicing pathway. Nat Neurosci 18, 817–825 (2015). https://doi.org/10.1038/nn.4019

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4019

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