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.

Astrocyte scar formation aids central nervous system axon regeneration

This article has been updated

Abstract

Transected axons fail to regrow in the mature central nervous system. Astrocytic scars are widely regarded as causal in this failure. Here, using three genetically targeted loss-of-function manipulations in adult mice, we show that preventing astrocyte scar formation, attenuating scar-forming astrocytes, or ablating chronic astrocytic scars all failed to result in spontaneous regrowth of transected corticospinal, sensory or serotonergic axons through severe spinal cord injury (SCI) lesions. By contrast, sustained local delivery via hydrogel depots of required axon-specific growth factors not present in SCI lesions, plus growth-activating priming injuries, stimulated robust, laminin-dependent sensory axon regrowth past scar-forming astrocytes and inhibitory molecules in SCI lesions. Preventing astrocytic scar formation significantly reduced this stimulated axon regrowth. RNA sequencing revealed that astrocytes and non-astrocyte cells in SCI lesions express multiple axon-growth-supporting molecules. Our findings show that contrary to the prevailing dogma, astrocyte scar formation aids rather than prevents central nervous system axon regeneration.

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: Preventing astrocytic scar formation does not lead to spontaneous regrowth of CST, AST or 5HT axons after SCI.
Figure 2: No spontaneous regrowth of CST, AST or 5HT axons after ablating chronic astrocyte scars.
Figure 3: CSPG production by non-astrocyte cells in SCI lesions after ablation or attenuation of astrocytic scars.
Figure 4: Genomic dissection of astrocytes and non-astrocyte cells in SCI lesions.
Figure 5: Robust regrowth of AST axons can be stimulated after wild-type SCI and is significantly attenuated by preventing astrocytic scar formation.

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Raw and normalized genomic data have been deposited in the NCBI Gene Expression Omnibus and are accessible through accession number GSE76097 and via a searchable, open-access website https://astrocyte.rnaseq.sofroniewlab.neurobio.ucla.edu

Change history

  • 13 April 2016

    Figure 5b and c were corrected to remove duplication of the ‘LC’ label in the bottom panels.

References

  1. Ramón y Cajal, S. Degeneration and Regeneration of the Nervous System (Oxford Univ. Press, 1928)

  2. Sun, F. et al. Sustained axon regeneration induced by co-deletion of PTEN and SOCS3. Nature 480, 372–375 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Liu, K., Tedeschi, A., Park, K. K. & He, Z. Neuronal intrinsic mechanisms of axon regeneration. Annu. Rev. Neurosci. 34, 131–152 (2011)

    Article  PubMed  CAS  Google Scholar 

  4. Richardson, P. M., McGuinness, U. M. & Aguayo, A. J. Axons from CNS neurons regenerate into PNS grafts. Nature 284, 264–265 (1980)

    Article  ADS  CAS  PubMed  Google Scholar 

  5. David, S. & Aguayo, A. J. Axonal elongation into peripheral nervous system “bridges” after central nervous system injury in adult rats. Science 214, 931–933 (1981)

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Schwab, M. E. Functions of Nogo proteins and their receptors in the nervous system. Nature Rev. Neurosci. 11, 799–811 (2010)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  8. Klapka, N. & Muller, H. W. Collagen matrix in spinal cord injury. J. Neurotrauma 23, 422–435 (2006)

    Article  PubMed  Google Scholar 

  9. Silver, J. & Miller, J. H. Regeneration beyond the glial scar. Nature Rev. Neurosci. 5, 146–156 (2004)

    Article  CAS  Google Scholar 

  10. Windle, W. F., Clemente, C. D. & Chambers, W. W. Inhibition of formation of a glial barrier as a means of permitting a peripheral nerve to grow into the brain. J. Comp. Neurol. 96, 359–369 (1952)

    Article  CAS  PubMed  Google Scholar 

  11. Windle, W. F. Regeneration of axons in the vertebrate central nervous system. Physiol. Rev. 36, 427–440 (1956)

    Article  CAS  PubMed  Google Scholar 

  12. Liuzzi, F. J. & Lasek, R. J. Astrocytes block axonal regeneration in mammals by activating the physiological stop pathway. Science 237, 642–645 (1987)

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Burda, J. E. & Sofroniew, M. V. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron 81, 229–248 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sofroniew, M. V. Astrocyte barriers to neurotoxic inflammation. Nature Rev. Neurosci. 16, 249–263 (2015)

    Article  CAS  Google Scholar 

  15. Bush, T. G. et al. Leukocyte infiltration, neuronal degeneration and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron 23, 297–308 (1999)

    Article  CAS  PubMed  Google Scholar 

  16. Faulkner, J. R. et al. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J. Neurosci. 24, 2143–2155 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Herrmann, J. E. et al. STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J. Neurosci. 28, 7231–7243 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wanner, I. B. et al. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J. Neurosci. 33, 12870–12886 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee, J. K. et al. Combined genetic attenuation of myelin and semaphorin-mediated growth inhibition is insufficient to promote serotonergic axon regeneration. J. Neurosci. 30, 10899–10904 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hawthorne, A. L. et al. The unusual response of serotonergic neurons after CNS injury: lack of axonal dieback and enhanced sprouting within the inhibitory environment of the glial scar. J. Neurosci. 31, 5605–5616 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nature Methods 2, 419–426 (2005)

    Article  CAS  PubMed  Google Scholar 

  22. Avnur, Z. & Geiger, B. Immunocytochemical localization of native chondroitin-sulfate in tissues and cultured cells using specific monoclonal antibody. Cell 38, 811–822 (1984)

    Article  CAS  PubMed  Google Scholar 

  23. Mikami, T. & Kitagawa, H. Biosynthesis and function of chondroitin sulfate. Biochim. Biophys. Acta 1830, 4719–4733 (2013)

    Article  CAS  PubMed  Google Scholar 

  24. Mironova, Y. A. & Giger, R. J. Where no synapses go: gatekeepers of circuit remodeling and synaptic strength. Trends Neurosci. 36, 363–373 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lin, A. C. & Holt, C. E. Local translation and directional steering in axons. EMBO J. 26, 3729–3736 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sanz, E. et al. Cell-type-specific isolation of ribosome-associated mRNA from complex tissues. Proc. Natl Acad. Sci. USA 106, 13939–13944 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zamanian, J. L. et al. Genomic analysis of reactive astrogliosis. J. Neurosci. 32, 6391–6410 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lang, B. T. et al. Modulation of the proteoglycan receptor PTPsigma promotes recovery after spinal cord injury. Nature 518, 404–408 (2015)

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Yamaguchi, Y. Lecticans: organizers of the brain extracellular matrix. Cell. Mol. Life Sci. 57, 276–289 (2000)

    Article  CAS  PubMed  Google Scholar 

  30. Miller, G. M. & Hsieh-Wilson, L. C. Sugar-dependentmodulation of neuronal development, regeneration, and plasticity by chondroitin sulfate proteoglycans. Exp. Neurol. 274, 115–125 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Goldberg, J. L. et al. Retinal ganglion cells do not extend axons by default: promotion by neurotrophic signaling and electrical activity. Neuron 33, 689–702 (2002)

    Article  CAS  PubMed  Google Scholar 

  32. Richardson, P. M. & Issa, V. M. Peripheral injury enhances central regeneration of primary sensory neurones. Nature 309, 791–793 (1984)

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Neumann, S. & Woolf, C. J. Regeneration of dorsal column fibers into and beyond the lesion site following adult spinal cord injury. Neuron 23, 83–91 (1999)

    Article  CAS  PubMed  Google Scholar 

  34. Omura, T. et al. Robust Axonal regeneration occurs in the injured CAST/Ei mouse CNS. Neuron 86, 1215–1227 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Alto, L. T. et al. Chemotropic guidance facilitates axonal regeneration and synapse formation after spinal cord injury. Nature Neurosci. 12, 1106–1113 (2009)

    Article  CAS  PubMed  Google Scholar 

  36. Plantman, S. et al. Integrin-laminin interactions controlling neurite outgrowth from adult DRG neurons in vitro. Mol. Cell. Neurosci. 39, 50–62 (2008)

    Article  CAS  PubMed  Google Scholar 

  37. Nowak, A. P. et al. Rapidly recovering hydrogel scaffolds from self-assembling diblock copolypeptide amphiphiles. Nature 417, 424–428 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  38. Yang, C. Y. et al. Biocompatibility of amphiphilic diblock copolypeptide hydrogels in the central nervous system. Biomaterials 30, 2881–2898 (2009)

    Article  CAS  PubMed  Google Scholar 

  39. Song, B. et al. Sustained local delivery of bioactive nerve growth factor in the central nervous system via tunable diblock copolypeptide hydrogel depots. Biomaterials 33, 9105–9116 (2012)

    Article  CAS  PubMed  Google Scholar 

  40. Tuszynski, M. H. & Steward, O. Concepts and methods for the study of axonal regeneration in the CNS. Neuron 74, 777–791 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Brosius Lutz, A. & Barres, B. A. Contrasting the glial response to axon injury in the central and peripheral nervous systems. Dev. Cell 28, 7–17 (2014)

    Article  CAS  PubMed  Google Scholar 

  42. Mason, C. A., Edmondson, J. C. & Hatten, M. E. The extending astroglial process: development of glial cell shape, the growing tip, and interactions with neurons. J. Neurosci. 8, 3124–3134 (1988)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kawaja, M. D. & Gage, F. H. Reactive astrocytes are substrates for the growth of adult CNS axons in the presence of elevated levels of nerve growth factor. Neuron 7, 1019–1030 (1991)

    Article  CAS  PubMed  Google Scholar 

  44. Zukor, K. et al. Short hairpin RNA against PTEN enhances regenerative growth of corticospinal tract axons after spinal cord injury. J. Neurosci. 33, 15350–15361 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Shih, C. H., Lacagnina, M., Leuer-Bisciotti, K. & Proschel, C. Astroglial-derived periostin promotes axonal regeneration after spinal cord injury. J. Neurosci. 34, 2438–2443 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhang, S. et al. Thermoresponsive copolypeptide hydrogel vehicles for CNS cell delivery. ACS Biomater. Sci. Eng . 1, 705–717 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ruschel, J. et al. Systemic administration of epothilone B promotes axon regeneration after spinal cord injury. Science 348, 347–352 (2015)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cafferty, W. B., McGee, A. W. & Strittmatter, S. M. Axonal growth therapeutics: regeneration or sprouting or plasticity ? Trends Neurosci. 31, 215–220 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bush, T. G. et al. Fulminant jejuno-ileitis following ablation of enteric glia in adult transgenic mice. Cell 93, 189–201 (1998)

    Article  CAS  PubMed  Google Scholar 

  50. Takeda, K. et al. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation and characterization of T cell-specific Stat3-deficient mice. J. Immunol. 161, 4652–4660 (1998)

    CAS  PubMed  Google Scholar 

  51. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nature Neurosci. 13, 133–140 (2010)

    Article  CAS  PubMed  Google Scholar 

  52. Zhang, S. et al. Tunable diblock copolypeptide hydrogel depots for local delivery of hydrophobic molecules in healthy and injured central nervous system. Biomaterials 35, 1989–2000 (2014)

    Article  CAS  PubMed  Google Scholar 

  53. Shigetomi, E. et al. Imaging calcium microdomains within entire astrocyte territories and endfeet with GCaMPs expressed using adeno-associated viruses. J. Gen. Physiol. 141, 633–647 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Jiang, R., Haustein, M. D., Sofroniew, M. V. & Khakh, B. S. Imaging intracellular Ca2 +signals in striatal astrocytes from adult mice using genetically-encoded calcium indicators. J. Vis. Exp . 93, e51972 (2014)

    Google Scholar 

  55. Tong, X. et al. Astrocyte Kir4.1 ion channel deficits contribute to neuronal dysfunction in Huntington’s disease model mice. Nature Neurosci. 17, 694–703 (2014)

    Article  CAS  PubMed  Google Scholar 

  56. Kang, S. H. et al. Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nature Neurosci. 16, 571–579 (2013)

    Article  CAS  PubMed  Google Scholar 

  57. Faul, F., Erdfelder, E., Lang, A. G. & Buchner, A. G. *Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behav. Res. Methods 39, 175–191 (2007)

    Article  PubMed  Google Scholar 

  58. Romero-Calvo, I. et al. Reversible Ponceau staining as a loading control alternative to actin in western blots. Anal. Biochem. 401, 318–320 (2010)

    Article  CAS  PubMed  Google Scholar 

  59. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013)

    Article  CAS  PubMed  Google Scholar 

  60. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015)

    Article  CAS  PubMed  Google Scholar 

  61. 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 

  62. Friedlander, D. R. et al. The neuronal chondroitin sulfate proteoglycan neurocan binds to the neural cell adhesion molecules Ng-CAM/L1/NILE and N-CAM, and inhibits neuronal adhesion and neurite outgrowth. J. Cell Biol. 125, 669–680 (1994)

    Article  CAS  PubMed  Google Scholar 

  63. Sango, K. et al. Phosphacan and neurocan are repulsive substrata for adhesion and neurite extension of adult rat dorsal root ganglion neurons in vitro. Exp. Neurol. 182, 1–11 (2003)

    Article  CAS  PubMed  Google Scholar 

  64. Hurtado, A., Podinin, H., Oudega, M. & Grimpe, B. Deoxyribozyme-mediated knockdown of xylosyltransferase-1 mRNA promotes axon growth in the adult rat spinal cord. Brain 131, 2596–2605 (2008)

    Article  PubMed  Google Scholar 

  65. Becker, C. G., Schweitzer, J., Feldner, J., Becker, T. & Schachner, M. Tenascin-R as a repellent guidance molecule for developing optic axons in zebrafish. J. Neurosci. 23, 6232–6237 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Dickson, B. J. Molecular mechanisms of axon guidance. Science 298, 1959–1964 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  67. Masuda, T. et al. Netrin-1 acts as a repulsive guidance cue for sensory axonal projections toward the spinal cord. J. Neurosci. 28, 10380–10385 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Winberg, M. L. et al. Plexin A is a neuronal semaphorin receptor that controls axon guidance. Cell 95, 903–916 (1998)

    Article  CAS  PubMed  Google Scholar 

  69. Hu, H., Marton, T. F. & Goodman, C. S. Plexin B mediates axon guidance in Drosophila by simultaneously inhibiting active Rac and enhancing RhoA signaling. Neuron 32, 39–51 (2001)

    Article  CAS  PubMed  Google Scholar 

  70. He, Z. & Tessier-Lavigne, M. Neuropilin is a receptor for the axonal chemorepellent Semaphorin III. Cell 90, 739–751 (1997)

    Article  CAS  PubMed  Google Scholar 

  71. Lu, X. et al. The netrin receptor UNC5B mediates guidance events controlling morphogenesis of the vascular system. Nature 432, 179–186 (2004)

    Article  ADS  CAS  PubMed  Google Scholar 

  72. Keino-Masu, K. et al. Deleted in Colorectal Cancer (DCC) encodes a netrin receptor. Cell 87, 175–185 (1996)

    Article  CAS  PubMed  Google Scholar 

  73. Ahmed, G. et al. Draxin inhibits axonal outgrowth through the netrin receptor DCC. J. Neurosci. 31, 14018–14023 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Rajagopalan, S. et al. Neogenin mediates the action of repulsive guidance molecule. Nature Cell Biol. 6, 756–762 (2004)

    Article  CAS  PubMed  Google Scholar 

  75. Monnier, P. P. et al. RGM is a repulsive guidance molecule for retinal axons. Nature 419, 392–395 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  76. Kajiwara, Y., Buxbaum, J. D. & Grice, D. E. SLITRK1 binds 14-3-3 and regulates neurite outgrowth in a phosphorylation-dependent manner. Biol. Psychiatry 66, 918–925 (2009)

    Article  CAS  PubMed  Google Scholar 

  77. Islam, S. M. et al. Draxin, a repulsive guidance protein for spinal cord and forebrain commissures. Science 323, 388–393 (2009)

    Article  ADS  CAS  PubMed  Google Scholar 

  78. Yang, Z. et al. NG2 glial cells provide a favorable substrate for growing axons. J. Neurosci. 26, 3829–3839 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Hossain-Ibrahim, M. K., Rezajooi, K., Stallcup, W. B., Lieberman, A. R. & Anderson, P. N. Analysis of axonal regeneration in the central and peripheral nervous systems of the NG2-deficient mouse. BMC Neurosci . 8, 80 (2007)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Lu, P., Jones, L. L. & Tuszynski, M. H. Axon regeneration through scars and into sites of chronic spinal cord injury. Exp. Neurol. 203, 8–21 (2007)

    Article  CAS  PubMed  Google Scholar 

  81. Busch, S. A. et al. Adult NG2 + cells are permissive to neurite outgrowth and stabilize sensory axons during macrophage-induced axonal dieback after spinal cord injury. J. Neurosci. 30, 255–265 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Nakanishi, K. et al. Identification of neurite outgrowth-promoting domains of neuroglycan C, a brain-specific chondroitin sulfate proteoglycan, and involvement of phosphatidylinositol 3-kinase and protein kinase C signaling pathways in neuritogenesis. J. Biol. Chem. 281, 24970–24978 (2006)

    Article  CAS  PubMed  Google Scholar 

  83. Götz, B. et al. Tenascin-C contains distinct adhesive, anti-adhesive, and neurite outgrowth promoting sites for neurons. J. Cell Biol. 132, 681–699 (1996)

    Article  PubMed  Google Scholar 

  84. Andrews, M. R. et al. Alpha9 integrin promotes neurite outgrowth on tenascin-C and enhances sensory axon regeneration. J. Neurosci. 29, 5546–5557 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Edwards, T. J. & Hammarlund, M. Syndecan promotes axon regeneration by stabilizing growth cone migration. Cell Rep . 8, 272–283 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Farhy Tselnicker, I., Boisvert, M. M. & Allen, N. J. The role of neuronal versus astrocyte-derived heparan sulfate proteoglycans in brain development and injury. Biochem. Soc. Trans. 42, 1263–1269 (2014)

    Article  PubMed  CAS  Google Scholar 

  87. Lu, P., Jones, L. L. & Tuszynski, M. H. BDNF-expressing marrow stromal cells support extensive axonal growth at sites of spinal cord injury. Exp. Neurol. 191, 344–360 (2005)

    Article  CAS  PubMed  Google Scholar 

  88. Grill, R., Murai, K., Blesch, A. & Tuszynski, M. H. Cellular delivery of neurotrophin-3 promotes coricospinal axonal growth and partial functional recovery after spinal cord injury. J. Neurosci. 17, 5560–5572 (1997)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Blesch, A. & Tuszynski, M. H. Cellular GDNF delivery promotes growth of motor and dorsal column sensory axons after partial and complete spinal cord transections and induces remyelination. J. Comp. Neurol. 467, 403–417 (2003)

    Article  CAS  PubMed  Google Scholar 

  90. Blesch, A. et al. Leukemia inhibitory factor augments neurotrophin expression and corticospinal axon growth after adult CNS injury. J. Neurosci. 19, 3556–3566 (1999)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Cafferty, W. B. et al. Leukemia inhibitory factor determines the growth status of injured adult sensory neurons. J. Neurosci. 21, 7161–7170 (2001)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Müller, A., Hauk, T. G. & Fischer, D. Astrocyte-derived CNTF switches mature RGCs to a regenerative state following inflammatory stimulation. Brain 130, 3308–3320 (2007)

    Article  PubMed  Google Scholar 

  93. Ozdinler, P. H. & Macklis, J. D. IGF-I specifically enhances axon outgrowth of corticospinal motor neurons. Nature Neurosci. 9, 1371–1381 (2006)

    Article  PubMed  CAS  Google Scholar 

  94. Szebenyi, G. et al. Fibroblast growth factor-2 promotes axon branching of cortical neurons by influencing morphology and behavior of the primary growth cone. J. Neurosci. 21, 3932–3941 (2001)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. White, R. E., Yin, F. Q. & Jakeman, L. B. TGF-α increases astrocyte invasion and promotes axonal growth into the lesion following spinal cord injury in mice. Exp. Neurol. 214, 10–24 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Tom, V. J., Doller, C. M., Malouf, A. T. & Silver, J. Astrocyte-associated fibronectin is critical for axonal regeneration in adult white matter. J. Neurosci. 24, 9282–9290 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Qin, J., Liang, J. & Ding, M. Perlecan antagonizes collagen IV and ADAMTS9/GON-1 in restricting the growth of presynaptic boutons. J. Neurosci. 34, 10311–10324 (2014)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Hill, J. J., Jin, K., Mao, X. O., Xie, L. & Greenberg, D. A. Intracerebral chondroitinase ABC and heparan sulfate proteoglycan glypican improve outcome from chronic stroke in rats. Proc. Natl Acad. Sci. USA 109, 9155–9160 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  99. Minor, K. et al. Decorin promotes robust axon growth on inhibitory CSPGs and myelin via a direct effect on neurons. Neurobiol. Dis. 32, 88–95 (2008)

    Article  CAS  PubMed  Google Scholar 

  100. Horie, H. et al. Galectin-1 regulates initial axonal growth in peripheral nerves after axotomy. J. Neurosci. 19, 9964–9974 (1999)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Walsh, F. S. & Doherty, P. Neural cell adhesion molecules of the immunoglobulin superfamily: role in axon growth and guidance. Annu. Rev. Cell Dev. Biol. 13, 425–456 (1997)

    Article  CAS  PubMed  Google Scholar 

  102. Malin, D. et al. The extracellular-matrix protein matrilin 2 participates in peripheral nerve regeneration. J. Cell Sci. 122, 995–1004 (2009)

    Article  CAS  PubMed  Google Scholar 

  103. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. W. Bergles for the NG2 antibody, and the Microscopy Core Resource of the UCLA Broad Stem Cell Research Center-CIRM Laboratory. This work was supported by the US National Institutes of Health (NS057624 and NS084030 to M.V.S.; P30 NS062691 to G.C. and NS060677, MH099559A, MH104069 to B.S.K.), and the Dr. Miriam and Sheldon G. Adelson Medical Foundation (M.V.S. and T.J.D.), and Wings for Life (M.V.S.).

Author information

Authors and Affiliations

Authors

Contributions

M.A.A., J.E.B., B.S.K., T.J.D. and M.V.S. designed experiments; M.A.A., J.E.B., Y.R. and Y.A. conducted experiments; M.A.A., J.E.B., Y.A., T.M.O., R.K., G.C. and M.V.S. analysed data. M.A.A., J.E.B., T.M.O., B.S.K, T.J.D. and M.V.S. prepared the manuscript.

Corresponding author

Correspondence to Michael V. Sofroniew.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 SCI model schematic, locomotor behavioural effects, and AAV vector targeting specificity and effects.

a, Schematic of severe lateral crush SCI at thoracic level T10 that generates a large lesion core (LC) of non-neural tissue surrounded by an astrocytic scar (AS) and completely transects descending and ascending axons. b, Open field hindlimb locomotor score at various times after SCI assessed using a 5-point scale where 5 is normal and 0 is no movement of any kind17. No significant differences were observed among any of the experimental groups at any time point. n = 6 mice at all time points P > 0.5 (ANOVA with Newman–Keuls post hoc analysis). WT, wild type. c, Horizontal sections through a severe SCI lesion of a representative tdTomato (tdT) reporter mouse51 injected with an AAV vector with a minimal Gfap promoter regulating Cre (AAV2/5-GfaABC1D-Cre) into the lesion at two weeks after SCI and perfused at three weeks. tdTomato labelling demonstrates that this AAV2/5-GfaABC1D-Cre efficiently and specifically targets GFAP-positive astrocytes. In this mouse, the amount AAV2/5-GfaABC1D-Cre injected was intentionally titrated on the basis of previous trial and error to target primarily the astrocytic scar border in an approximately 500 μm zone immediately abutting the SCI lesion core. High-magnification analysis of individual fluorescence channels stained for tdTomato plus various cell markers shows the specificity of Cre activity targeting to cells expressing the astrocyte marker, GFAP, but not to cells expressing either the neuronal marker, NeuN, or the mature oligodendrocyte marker, GSTπ. AAV2/5-GfaABC1D-Cre was prepared using a previously described and well-characterized cloning strategy53,54,55. d, Open field hindlimb locomotor scores at various times after SCI. There was no difference in scores of control mice and loxP-STOP-loxP-DTR (diphtheria toxin receptor) mice that received AAV2/5-GfaABC1D-Cre before injections of diphtheria toxin (DT). Five weeks after DT injections, loxP-DTR mice that received AAV2/5- GfaABC1D-Cre exhibited a slightly, but significantly, lower locomotor score. Hindlimb locomotion was assessed using a 5-point scale where 5 is normal and 0 is no movement of any kind17. n = 6 mice per group; *P < 0.05 versus wild-type (ANOVA with Newman–Keuls). e, GFAP immunohistochemistry of a sagittal section after ablation of a chronic astrocytic scar plus adjacent astrocytes. DT was administered to a transgenic mouse expressing DTR targeted selectively to astrocytes around a severe SCI. In this case, the amount of AAV2/5-GfaABC1D-Cre injected was titrated to target not only primarily the astrocytic scar border but also adjacent astrocytes spread over approximately 2 mm on either side of the centre (Cn) of the SCI lesion core (LC). Note the profound degeneration of neural tissue resulting from the selective ablation of the chronic astrocytic scar plus adjacent astrocytes after SCI.

Source data

Extended Data Figure 2 Single-channel CSPG and GFAP immunofluorescence and stained area quantification.

a, Individual fluorescence channels of CS56 and GFAP immunohistochemistry from horizontal sections of uninjured mice and at two weeks after severe SCI shown in Fig. 3b. Sections are taken from wild-type (WT) mice and mice with transgenic ablation (TK+GCV) or attenuation (STAT3-CKO) of astrocytic scar formation. b, Example of black and white thresholding of single channels of immunofluorescence staining for image analysis to quantify (using NIH Image J software) the amount of CSPG- or GFAP-stained area in different tissue compartments in SCI lesions. Boxes denote areas quantified to obtain values for lesion core (LC) and grey (g) or white (w) matter in astrocytic scar (AS) or equivalent regions in uninjured tissue. Graphs show percentage of areas (means ± s.e.m.) stained for CSPG or GFAP determined using ImageJ. n = 4 (wild type mice); n = 6 (TK+GCV and STAT3-CKO mice); #P < 0.05 versus uninjured white matter; *P < 0.05 versus uninjured grey matter in same experimental group (ANOVA with Newman–Keuls); ^P < 0.05 versus equivalent anatomical region in wild-type (ANOVA with Newman–Keuls).

Source data

Extended Data Figure 3 Specificity of haemagglutinin targeting to astrocytes and enrichment of haemagglutinin immunoprecipitation for astrocyte-specific RNA transcripts.

a, Individual fluorescence channels of immunohistochemistry for transgenically targeted haemagglutinin (HA) plus various cell markers showing the specificity of HA targeting to cells expressing the astrocyte marker, GFAP, and not to cells expressing either the neuronal marker, NeuN, or the mature oligodendrocyte marker, GSTπ, in uninjured grey and white matter and in astrocytic scars at 2 weeks after SCI. b, CNS-cell-type-specific gene transcript enrichment of ribosome-associated mRNA (ramRNA) isolated from wild-type (WT) uninjured spinal cord by HA immunoprecipitation (HA-IP). Differential expression analysis by RNA-seq indicates significant enrichment (red) for astrocyte-specific gene transcripts, and de-enrichment (green) for gene transcripts enriched in other CNS cell types, FDR < 0.1. A log2 scale is used so that positive and negative differences are directly comparable. The mean numerical enrichment of three quintessential astrocyte genes, Gfap, Aldh1l1 and Aqp4, is 25-fold greater in HA samples than in flow-through samples. c, Gene transcript enrichment of HA-IP ramRNA relative to P7 mouse primary cortical astrocytes103. Of the 200 most highly expressed genes previously described103 for post-natal mouse cortical astrocytes, 71.5% (red line) are at least fourfold enriched (blue line) in HA-IP ramRNA isolated from uninjured spinal cord relative to flow through RNA from non-astrocyte cells. d, Pearson correlation plots of total normalized RNA-seq reads from individual biological replicates for each treatment condition. Correlation colouring indicates little (white) to high (red) similarity. n = 4 mice each for uninjured controls and wild-type SCI (SCI-WT); n = 3 mice for STAT3-CKO SCI (SCI-STAT3). FDR < 0.1 for differential expression and enrichment analysis. Raw and normalized data have been deposited in the NCBI Gene Expression Omnibus and are accessible through GEO Series accession number GSE76097 and via a searchable, open-access website https://astrocyte.rnaseq.sofroniewlab.neurobio.ucla.edu.

Extended Data Figure 4 Comparison of genomic data from astrocytes and non-astrocyte cells from WT and STAT3-CKO mice after SCI.

a, Heat maps depicting all significantly differentially expressed genes (DEG), as determined by RNA-seq, for wild-type (WT) and STAT3-CKO astrocytes and non-astrocytes from independent biological replicates two weeks after SCI relative to uninjured wild-type control. Red upregulated, green downregulated. b, Total numbers and Venn diagrams of significant DEGs in wild-type and STAT3-CKO astrocytes and non-astrocytes two weeks after SCI relative to uninjured control. Red and green numerical values indicate significantly upregulated and downregulated genes, respectively. c, Comparison of altered gene expression in our SCI-reactive astrocytes and previously reported forebrain stroke-reactive astrocytes27. Of the 200 most highly elevated genes in forebrain astrocytes one week following stroke27, 58.5% (red line) are also significantly elevated in astrocytes after SCI, relative to uninjured. d, Comparison of expression by wild-type SCI and STAT3-CKO SCI reactive astrocytes of a selected cross-section of genes that are highly regulated after SCI by wild-type reactive astrocytes. Many of the regulated genes exhibit changes that are expected and implicated in wild-type reactive astrogliosis mechanisms and roles, and some of the changes appear to be newly identified in this context. Note that many of the genes are not regulated or exhibit attenuated changes in STAT3-CKO SCI astrocytes. n = 4 mice each for uninjured and wild-type SCI; n = 3 mice for STAT3-CKO SCI (SCI-STAT3). FDR < 0.1 for differential expression and enrichment analysis.

Extended Data Figure 5 Immunohistochemistry of specific CSPGs.

a, Absence of aggrecan (ACAN) production by scar-forming astrocytes. Images show individual fluorescence channels of ACAN and GFAP immunohistochemistry from horizontal sections two weeks after severe SCI in a representative wild-type (WT) mouse. Boxes denote areas of astrocytic scar (AS) or uninjured tissue (Uninj) shown at higher magnification. Note that ACAN is: (i) heavily present in the perineuronal nets that surround neurons in uninjured tissue; (ii) almost absent from astrocytic scar and lesion core (LC); and (iii) not detectably produced by newly generated scar-forming astrocytes (arrows). b, Brevican (BCAN) production by scar-forming astrocytes and non-astrocyte cells. Images show individual fluorescence channels of BCAN and GFAP immunohistochemistry from horizontal sections two weeks after severe SCI, in wild-type mice and mice with transgenic ablation (TK+GCV) or attenuation (STAT3-CKO) of astrocytic scar formation. Note that BCAN is produced both by GFAP-positive scar-forming astrocytes (arrowheads) and by non-astrocyte cells (arrows).

Extended Data Figure 6 Immunohistochemistry of specific CSPGs.

a, Neurocan (NCAN) production by scar-forming astrocytes and non-astrocyte cells. Images show individual fluorescence channels of NCAN and GFAP immunohistochemistry from horizontal sections two weeks after severe SCI, in a representative wild-type (WT) mouse. Box denotes area of lesion core (LC) and astrocytic scar (AS) shown at higher magnification. Note that NCAN is produced both by GFAP-positive scar-forming astrocytes and by non-astrocyte cells (arrows) in the lesion core. b, NG2 (CSPG4) production by newly proliferated scar-forming astrocytes. Images show individual channels and various combinations of immunofluorescence staining for NG2, GFAP, tdTomato (tdT), BrdU (proliferation marker) and DAPI showing astrocytes in a mature SCI scar. The images are representative of findings from tdTomato-reporter mice51 injected with AAV2/5-GfaABC1D-Cre vector53 into multiple sites of the uninjured spinal cord to label mature astrocytes. Three weeks after AAV2/5-GfaABC1D-Cre injection, the mice received a severe SCI and were administered BrdU from days 2–7 after SCI. The mice were perfused after two weeks after SCI. Images comparing individual fluorescence channels show that astrocytes labelled 1 and 3: (i) incorporated BrdU and thus are newly proliferated after SCI; (ii) express the tdTomato reporter; (iii) express GFAP, the prototypical marker of reactive and scar-forming astrocytes; and (iv) express NG2 both intracellularly and along their cell surfaces. In contrast, astrocyte number 2 is also BrdU-labelled and expresses both tdTomato and GFAP, but does not appear to express detectable levels of NG2. c, CSPG5 (Neuroglycan C) production by scar-forming astrocytes. Images show individual channels and various combinations of immunofluorescence staining for CSPG5 or GFAP. Note that CSPG5 is present within and along the processes of GFAP-positive scar-forming astrocytes (arrows).

Extended Data Figure 7 Specificity and effects of treatments to stimulate AST axon regrowth after SCI.

a, BDNF and NT3 treatment does not alter the appearance or density of astrocyte scars in wild-type (WT) or STAT3-CKO mice. Images show horizontal sections of mice at two weeks after SCI or after SCI followed by delayed injection of hydrogel only (as a control) or hydrogel releasing NT3 and BDNF. Top images show GFAP immunofluorescence; boxed area denotes size of areas taken from multiple locations in the astrocytic scar (AS) for GFAP area quantification shown in graph. n = 5 mice per group; NS, P > 0.05 (ANOVA with Newman–Keuls). Bottom images show brightfield immunohistochemistry simultaneously of GFAP+TK to stain both astrocyte cell processes (GFAP) and cell bodies (TK) in mGFAP-TK transgenic mice for quantification of astrocyte cell numbers shown in graph. For these experiments the transgene-derived TK is used as a reporter protein that efficiently labels astrocyte cell bodies and thereby improves cell quantification18 and the mice were not given GCV. n = 4 mice per group; *P < 0.05 versus uninjured (ANOVA with Newman–Keuls); NS, P > 0.05 (ANOVA with Newman–Keuls). b, AST axon regrowth through scar-forming astrocytes and CSPGs in SCI lesions. Images show individual channels and various combinations of immunofluorescence staining for CTB, GFAP and CS56 to detect total CSPGs from a wild-type mouse after SCI followed by delayed injection of a hydrogel depot releasing NT3 and BDNF, shown as multichannel image in Fig. 5e. Arrows denote robust regrowth of many AST axons along, through and past scar-forming astrocytes into and through the lesion core. Note that the stimulated axons are regrowing through CSPG containing areas in the astrocyte scar and lesion core. Boxed area is shown at higher magnification in Extended Data Fig. 8. c, Graph shows numbers of AST axons at various distances past the proximal border of the astrocytic scar under different conditions. n = 5 per group; *P < 0.001 significant difference SCI+CL+BDNF+NT3 versus all other groups (ANOVA with post-hoc Newman–Keuls).

Source data

Extended Data Figure 8 AST axon regrowth through scar-forming astrocytes and CSPGs in SCI lesions.

a, b, Images show individual channels and various combinations of immunofluorescence staining for CTB, GFAP and CS56 to detect total CSPGs from a wild-type (WT) mouse after SCI followed by delayed injection of a hydrogel depot releasing NT3 and BDNF, shown as multichannel images in Fig. 5f, g. Arrows in a denote robust regrowth of many AST axons along, through and past scar-forming astrocytes into and through the lesion core; note that the stimulated axons are regrowing through CSPG containing areas in the astrocytic scar and lesion core. b, High-magnification orthogonal images of axons in three visual planes. Arrows in b denote AST regrowing axons tracking along CSPG-positive and GFAP-negative structures. Arrowheads in b denote AST axons tracking along GFAP-positive and CSPG-positive astrocyte processes, passing from one astrocyte process to another.

Extended Data Figure 9 AST axon regrowth in SCI lesions is dependent on laminin.

ag, Tract tracing of AST axons using CTB and laminin immunohistochemistry. ac, Same fields imaged for CTB alone (a1c1), or CTB plus laminin (a2c2). a, d, Intact gracile–cuneate tract (GCT). b, SCI only. c, f, SCI plus conditioning lesion (CL) plus hydrogel with growth factors. e, SCI plus conditioning lesion. g, SCI plus conditioning lesion plus hydrogel with growth factors and anti-CD29. dg, High-magnification orthogonal images of axons in three visual planes. Arrows indicate regrowing axons in direct contact with laminin. Arrowheads indicate axons not in direct contact with laminin in the intact GCT (d) or with anti-CD29 treatment (g). Note the difference in appearance of axons in the intact gracile–cuneate tract (GCT), which are independent of laminin, compared with regrowing axons in lesion core (LC), which track along laminin. h, Axon length per tissue volume (means ± s.e.m.) in intact GCT or in SCI lesions under different conditions. Intact GCT values were not included in ANOVA comparison of other 3 groups. i, Percentages (means ± s.e.m.) of AST axon length in direct contact with laminin under different conditions. n = 5 mice per group; *P < 0.001 (ANOVA with post-hoc Newman–Keuls); NS, not significant (ANOVA with post-hoc Newman–Keuls).

Source data

Extended Data Table 1 Axon growth inhibitory molecules and axon growth permissive molecules

Supplementary information

Supplementary Information

This file contains Supplementary Text. (PDF 101 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Anderson, M., Burda, J., Ren, Y. et al. Astrocyte scar formation aids central nervous system axon regeneration. Nature 532, 195–200 (2016). https://doi.org/10.1038/nature17623

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature17623

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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