Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Reports

tubule stabilization induces such


Systemic administration of epothilone B divergent effects. Moreover, Tax-
ol cannot be used for clinical
promotes axon regeneration after spinal CNS intervention because it does
not cross the blood-brain barrier
cord injury (10).
We aimed to target microtu-
bule stabilization in the injured
Jörg Ruschel,1 Farida Hellal,1*† Kevin C. Flynn,1*‡ Sebastian CNS in a clinically feasible way
Dupraz, * David A. Elliott, Andrea Tedeschi, Margaret Bates,
1 1 1 2 and to decipher its distinct cellu-
lar actions. We used epothilones,
Christopher Sliwinski, Gary Brook, Kristina Dobrindt,
3 4,5 6
a class of FDA approved blood-
Michael Peitz,6 Oliver Brüstle,6 Michael D. Norenberg,7 Armin brain barrier permeable micro-
Blesch,3 Norbert Weidner,3 Mary Bartlett Bunge,2 John L. Bixby,2 tubule stabilizing drugs (11).
Frank Bradke1§ Mass spectrometry confirmed
1
that after intraperitoneal (i.p.)
Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn,
2
Germany. The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 Northwest 14th injection in adult rats, epoB was
Terrace, Miami, FL33136, USA. 3Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstr. 200A, rapidly absorbed into the CNS
69118 Heidelberg, Germany. 4Institute for Neuropathology, RWTH Aachen University, Steinbergweg 20, 52074, Aachen, and remained at comparable
Germany. 5Jülich-Aachen Research Alliance–Translational Brain Medicine. 6Institute of Reconstructive Neurobiology, levels for 6 days (Fig. 1A). Rats
Life&Brain Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany.
7
Departments of Pathology, Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL 33101, i.p. injected with 0.75 mg/kg
USA.. body weight (BW) epoB at day 1
*These authors contributed equally to this work. and 15 post-injury showed in-
creased levels of detyrosinated
†Present address: Institute for Stroke and Vascular Dementia Research, University of Munich Medical Center, Max
and acetylated tubulin in lesion
Lebsche Platz 30, 81377 Munich, Germany.
site extracts 4 weeks after spinal
‡Present address: Department of Molecular Medicine, Max Planck Institute of Biochemistry, Am Klopferspitz 18, cord dorsal hemisection (Fig. 1B)
82152 Martinsried, Germany. indicating increased microtubule
§Corresponding author. E-mail: frank.bradke@dzne.de stability (12). The dosage used
presented no obvious adverse
After central nervous system (CNS) injury, inhibitory factors in the lesion
side effects, such as reduced an-
scar and poor axon growth potential prevent axon regeneration.
imal weight or decreased white
Microtubule stabilization reduces scarring and promotes axon growth.
blood cell counts (fig. S1).
However, the cellular mechanisms of this dual effect remain unclear. Here,
Fibrotic scar tissue rich in fi-
delayed systemic administration of a blood-brain barrier permeable
bronectin and laminin forms at
microtubule stabilizing drug, epothilone B (epoB), decreased scarring after
the lesion site after SCI in ro-
rodent spinal cord injury (SCI) by abrogating polarization and directed
dents (8) and humans (Fig. 1C;
migration of scar-forming fibroblasts. Conversely, epothilone B reactivated
table S1). This scar tissue poses a
neuronal polarization by inducing concerted microtubule polymerization
key impediment to regenerating
into the axon tip, which propelled axon growth through an inhibitory
axons, because it contains axon
environment. Together, these drug elicited effects promoted axon
growth inhibitory factors, includ-
regeneration and improved motor function after SCI. With recent clinical
ing chondroitin sulfate proteo-
approval, epothilones hold promise for clinical use after CNS injury.
glycans (CSPGs) (1, 8). Adult rats
systemically treated post-injury
An ideal treatment to induce axon regeneration in the in- with 0.75 mg/kg BW epoB showed a significant reduction of
jured CNS should reduce scarring (1) and growth inhibitory fibronectin (Fig. 1B) and of laminin-positive fibrotic scar
factors at the lesion site (2–4), reactivate the axon growth tissue even 4 weeks after dorsal hemisection (Fig. 1D and E).
potential (5) and be administrable as a medication after in- We found a comparable decrease of fibrotic scarring when
jury. Recently, a number of combinatorial approaches have epoB was locally delivered to the injury site via an intrathe-
led to axon regeneration (6, 7). These approaches, however, cal catheter (fig. S2) (8). Reduction of fibrotic scar tissue by
involve multiple drugs, enzymes and interventions render- systemic epoB administration was associated with a de-
ing clinical translation difficult. Moderate microtubule sta- crease of CSPGs (Fig. 1, D and F), including neurocan (Fig.
bilization by the anti-cancer drug Taxol promotes axon 1B) and NG2 (13), at the injury site (fig. S3). Astrogliosis and
regeneration by reducing fibrotic scarring and increasing lesion area were similar between treated and control ani-
axon growth (8, 9). However, it remains elusive how micro- mals (fig. S1) indicating that neuroprotective glial sealing of

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 1 / 10.1126/science.aaa2958


the injury site (14) was not affected by the treatment. tured neurons, we assessed its ability to promote axon re-
Scar reduction upon epoB treatment resulted neither generation after SCI. In vivo imaging of adult transgenic
from decreased cell proliferation nor from increased apop- mice, expressing green fluorescent protein (GFP) in spinal
tosis (fig. S4) but from a migratory defect of scar-forming cord dorsal column axons (23, 24), revealed that transected
meningeal fibroblasts (15). In wound healing assays, epoB axons of animals injected with 1.5 mg/kg BW epoB exhibited
inhibited migration of meningeal fibroblasts (Fig. 1, G and significantly fewer retraction bulbs (Fig. 3, C and D), re-
H, and movies S1 and S2) by changing their microtubular duced axonal dieback and increased regenerative growth
network. Control cells polarized by forming a leading edge (Fig. 3, C and E). Moreover, in adult mice, systemic and
enriched in stable detyrosinated microtubules and a trailing post-injury treatment with epoB, promoted axon regenera-
edge containing dynamic, tyrosinated microtubules (Fig. 1I), tion after complete dorsal column transection (Fig. 3, F and
both hallmarks of directed cell migration (16). In contrast, G).
epoB treated fibroblasts were round and nonpolar (Fig. 1J We then tested whether the treatment also promoted ax-
and fig. S5) with elevated levels of detyrosinated microtu- on regrowth of descending axons important for locomotion.
bules (Fig. 1K) distributed throughout the cell (Fig. 1J). Simi- In adult rats post-injury injected with 0.75 mg/kg BW epoB,
larly, systemic administration of epoB after dorsal we found a 3-fold increase of serotonergic fibers caudal to a
hemisection prevented the polarization of meningeal fibro- dorsal hemisection (Fig. 4, A and B). Increased serotonergic
blasts at the lesion site into a bipolar, migratory shape (Fig. innervation strongly correlates with recovery of motor func-
1L), which reduced scar formation (Fig. 1D and E). tion after SCI (25–27). Therefore, we asked whether the
In co-cultures of meningeal fibroblasts and postnatal treatment improves walking of adult rats that underwent a
cortical neurons, epoB treatment (1 nM) perturbed fibro- moderate, mid-thoracic spinal cord contusion, a clinically
blast polarization while enhancing axon growth (fig. S5). relevant SCI model (28). After contusion injury, epoB ad-
Moreover, epoB restored axon growth when these neurons ministration (0.75 mg/kg BW) reduced fibrotic scarring at
were confronted with the inhibitory molecules Nogo-A, the injury site (fig. S10) and promoted serotonergic axon
CSPGs or Semaphorin 3A (Fig. 2, A and B), which are abun- regrowth in the caudal spinal cord (Fig. 4, C and D). Moreo-
dant at the spinal cord lesion site (2–4, 17). In neurons ex- ver, epoB treatment increased stride length and gait regular-
pressing fluorescently-tagged microtubule plus-end binding ity, and reduced external rotation of the hind paws (fig. S11)
protein 3 (EB3-mCherry), which labels polymerizing micro- indicating improved walking balance and coordination. Ac-
tubules (18), epoB induced rapid and concerted microtubule cordingly, epoB treated animals showed a 50% reduction of
polymerization into the neurite tips (Fig. 2, C and D, and foot misplacements on the horizontal ladder compared to
movie S3) causing axon elongation despite inhibitory Nogo- injured controls (Fig. 4E and movies S5 and S6). These func-
A (Fig. 2E and movie S3). In accordance, low doses of the tional improvements were abrogated by pharmacological
microtubule destabilizing drug nocodazole abolished micro- ablation of serotonergic innervation (Fig. 4, D and E, and
tubule protrusion in neurites (Fig. 2, D and F) and abrogat- movies S7 and S8) (25).
ed the growth promoting effect of epoB (Fig. 2B). EpoB also The finding that the stabilization of microtubules inhib-
promoted axon growth of human cortical neurons under its cell division established the usage of systemic microtu-
growth permissive as well as non-permissive conditions (fig. bule stabilizing agents as a therapeutic standard for the
S6). In meningeal fibroblasts, however, epoB prevented mi- treatment of cancer (29). Here, at low doses, systemic ad-
crotubule polymerization toward the cell edges (Fig. 2G) ministration of the microtubule stabilizing agent epoB pro-
contrasting with the microtubule dynamics found in neu- moted functional recovery after SCI. Our approach differs
rons. This dichotomy was due to neuron-specific expression from other experimental regenerative paradigms (1–7) by
of the microtubule associated protein Tau (fig. S7), which pharmacologically focusing on a single molecular target, the
regulates microtubule dynamics, bundling, and binding of microtubules, yet overcoming multiple pathological obsta-
microtubule stabilizing agents (19, 20). In fibroblasts ectopi- cles. This is possible due to divergent effects of pharmaco-
cally expressing Tau, epoB induced an accumulation of logical microtubule stabilization on microtubule dynamics
bundled microtubules (fig. S8) that polymerized toward the and, hence the polarization of neurons and meningeal fi-
cell edge (Fig. 2, H and I, and movie S4), mimicking the ef- broblasts. This dual effect and the efficacy after systemic
fect observed in neurons. In turn, neurons depleted of Tau, and post-injury administration, give epothilones a promis-
by transfection with a plasmid encoding small hairpin (sh) ing translational perspective for treatment of the injured
RNA for tau (21), showed reduced microtubule polymeriza- CNS.
tion into the distal neurite when exposed to epoB (fig. S9). REFERENCES AND NOTES
Injured axons in the rodent and human CNS form dys- 1. N. Klapka, S. Hermanns, G. Straten, C. Masanneck, S. Duis, F. P. Hamers, D. Müller,
trophic retraction bulbs (Fig. 3, A to D, and table S2), a con- W. Zuschratter, H. W. Müller, Suppression of fibrous scarring in spinal cord injury
sequence of microtubule depolymerization and of rat promotes long-distance regeneration of corticospinal tract axons, rescue
of primary motoneurons in somatosensory cortex and significant functional
disorganization (Fig. 3, A and B) (22, 23). Because epoB in- recovery. Eur. J. Neurosci. 22, 3047–3058 (2005). Medline doi:10.1111/j.1460-
duced microtubule polymerization and axon growth in cul- 9568.2005.04495.x

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 2 / 10.1126/science.aaa2958


2. T. GrandPré, S. Li, S. M. Strittmatter, Nogo-66 receptor antagonist peptide 8320 (2005). Medline doi:10.1073/pnas.0408974102
promotes axonal regeneration. Nature 417, 547–551 (2002). Medline 21. H. Zempel, J. Luedtke, Y. Kumar, J. Biernat, H. Dawson, E. Mandelkow, E. M.
doi:10.1038/417547a Mandelkow, Amyloid-β oligomers induce synaptic damage via Tau-dependent
3. E. J. Bradbury, L. D. Moon, R. J. Popat, V. R. King, G. S. Bennett, P. N. Patel, J. W. microtubule severing by TTLL6 and spastin. EMBO J. 32, 2920–2937 (2013).
Fawcett, S. B. McMahon, Chondroitinase ABC promotes functional recovery Medline doi:10.1038/emboj.2013.207
after spinal cord injury. Nature 416, 636–640 (2002). Medline 22. F. Bradke, J. W. Fawcett, M. E. Spira, Assembly of a new growth cone after
doi:10.1038/416636a axotomy: The precursor to axon regeneration. Nat. Rev. Neurosci. 13, 183–193
4. L. Schnell, M. E. Schwab, Axonal regeneration in the rat spinal cord produced by (2012). Medline
an antibody against myelin-associated neurite growth inhibitors. Nature 343, 23. A. Ertürk, F. Hellal, J. Enes, F. Bradke, Disorganized microtubules underlie the
269–272 (1990). Medline doi:10.1038/343269a0 formation of retraction bulbs and the failure of axonal regeneration. J. Neurosci.
5. K. Liu, Y. Lu, J. K. Lee, R. Samara, R. Willenberg, I. Sears-Kraxberger, A. Tedeschi, 27, 9169–9180 (2007). Medline doi:10.1523/JNEUROSCI.0612-07.2007
K. K. Park, D. Jin, B. Cai, B. Xu, L. Connolly, O. Steward, B. Zheng, Z. He, PTEN 24. C. J. Laskowski, F. Bradke, In vivo imaging: A dynamic imaging approach to study
deletion enhances the regenerative ability of adult corticospinal neurons. Nat. spinal cord regeneration. Exp. Neurol. 242, 11–17 (2013). Medline
Neurosci. 13, 1075–1081 (2010). Medline doi:10.1038/nn.2603 doi:10.1016/j.expneurol.2012.07.007
6. W. J. Alilain, K. P. Horn, H. Hu, T. E. Dick, J. Silver, Functional regeneration of 25. S. Kaneko, A. Iwanami, M. Nakamura, A. Kishino, K. Kikuchi, S. Shibata, H. J.
respiratory pathways after spinal cord injury. Nature 475, 196–200 (2011). Okano, T. Ikegami, A. Moriya, O. Konishi, C. Nakayama, K. Kumagai, T. Kimura, Y.
Medline doi:10.1038/nature10199 Sato, Y. Goshima, M. Taniguchi, M. Ito, Z. He, Y. Toyama, H. Okano, A selective
7. P. Lu, Y. Wang, L. Graham, K. McHale, M. Gao, D. Wu, J. Brock, A. Blesch, E. S. Sema3A inhibitor enhances regenerative responses and functional recovery of
Rosenzweig, L. A. Havton, B. Zheng, J. M. Conner, M. Marsala, M. H. Tuszynski, the injured spinal cord. Nat. Med. 12, 1380–1389 (2006). Medline
Long-distance growth and connectivity of neural stem cells after severe spinal doi:10.1038/nm1505
cord injury. Cell 150, 1264–1273 (2012). Medline doi:10.1016/j.cell.2012.08.020 26. J. E. Kim, B. P. Liu, J. H. Park, S. M. Strittmatter, Nogo-66 receptor prevents
8. F. Hellal, A. Hurtado, J. Ruschel, K. C. Flynn, C. J. Laskowski, M. Umlauf, L. C. raphespinal and rubrospinal axon regeneration and limits functional recovery
Kapitein, D. Strikis, V. Lemmon, J. Bixby, C. C. Hoogenraad, F. Bradke, from spinal cord injury. Neuron 44, 439–451 (2004). Medline
Microtubule stabilization reduces scarring and causes axon regeneration after doi:10.1016/j.neuron.2004.10.015
spinal cord injury. Science 331, 928–931 (2011). Medline 27. U. Sławińska, K. Miazga, A. M. Cabaj, A. N. Leszczyńska, H. Majczyński, J. I. Nagy,
9. V. Sengottuvel, M. Leibinger, M. Pfreimer, A. Andreadaki, D. Fischer, Taxol L. M. Jordan, Grafting of fetal brainstem 5-HT neurons into the sublesional spinal
facilitates axon regeneration in the mature CNS. J. Neurosci. 31, 2688–2699 cord of paraplegic rats restores coordinated hindlimb locomotion. Exp. Neurol.
(2011). Medline doi:10.1523/JNEUROSCI.4885-10.2011 247, 572–581 (2013). Medline doi:10.1016/j.expneurol.2013.02.008
10. S. Fellner, B. Bauer, D. S. Miller, M. Schaffrik, M. Fankhänel, T. Spruss, G. 28. J. R. Wrathall, R. K. Pettegrew, F. Harvey, Spinal cord contusion in the rat:
Bernhardt, C. Graeff, L. Färber, H. Gschaidmeier, A. Buschauer, G. Fricker, Production of graded, reproducible, injury groups. Exp. Neurol. 88, 108–122
Transport of paclitaxel (Taxol) across the blood-brain barrier in vitro and in vivo. (1985). Medline doi:10.1016/0014-4886(85)90117-7
JCI 110, 1309–1318 (2002). Medline doi:10.1172/JCI0215451 29. C. Dumontet, M. A. Jordan, Microtubule-binding agents: A dynamic field of
11. C. Ballatore, K. R. Brunden, D. M. Huryn, J. Q. Trojanowski, V. M. Lee, A. B. Smith cancer therapeutics. Nat. Rev. Drug Discov. 9, 790–803 (2010). Medline
3rd, Microtubule stabilizing agents as potential treatment for Alzheimer’s doi:10.1038/nrd3253
disease and related neurodegenerative tauopathies. J. Med. Chem. 55, 8979– 30. S. W. Scheff, A. G. Rabchevsky, I. Fugaccia, J. A. Main, J. E. Lumpp Jr.,
8996 (2012). Medline doi:10.1021/jm301079z Experimental modeling of spinal cord injury: Characterization of a force-defined
12. C. Janke, J. C. Bulinski, Post-translational regulation of the microtubule injury device. J. Neurotrauma 20, 179–193 (2003). Medline
cytoskeleton: Mechanisms and functions. Nat. Rev. Mol. Cell Biol. 12, 773–786 doi:10.1089/08977150360547099
(2011). Medline doi:10.1038/nrm3227 31. M. Carta, T. Carlsson, D. Kirik, A. Björklund, Dopamine released from 5-HT
13. J. M. Cregg, M. A. DePaul, A. R. Filous, B. T. Lang, A. Tran, J. Silver, Functional terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats. Brain
regeneration beyond the glial scar. Exp. Neurol. 253, 197–207 (2014). Medline 130, 1819–1833 (2007). Medline doi:10.1093/brain/awm082
doi:10.1016/j.expneurol.2013.12.024 32. M. Bates, R. Puzis, M. Bunge, Preparation of spinal cord injured tissue for light
14. J. R. Faulkner, J. E. Herrmann, M. J. Woo, K. E. Tansey, N. B. Doan, M. V. and electron microscopy including preparation for immunostaining. Animal
Sofroniew, Reactive astrocytes protect tissue and preserve function after spinal Models of Movement Disorders 62, 381–399 (2011). doi:10.1007/978-1-61779-
cord injury. J. Neurosci. 24, 2143–2155 (2004). Medline 301-1_20
doi:10.1523/JNEUROSCI.3547-03.2004 33. A. Buss, K. Pech, B. A. Kakulas, D. Martin, J. Schoenen, J. Noth, G. A. Brook,
15. A. L. Carbonell, J. Boya, Ultrastructural study on meningeal regeneration and Growth-modulating molecules are associated with invading Schwann cells and
meningo-glial relationships after cerebral stab wound in the adult rat. Brain Res. not astrocytes in human traumatic spinal cord injury. Brain 130, 940–953
439, 337–344 (1988). Medline doi:10.1016/0006-8993(88)91491-6 (2007). Medline doi:10.1093/brain/awl374
16. G. G. Gundersen, J. C. Bulinski, Selective stabilization of microtubules oriented 34. J. C. Fleming, M. D. Norenberg, D. A. Ramsay, G. A. Dekaban, A. E. Marcillo, A. D.
toward the direction of cell migration. Proc. Natl. Acad. Sci. U.S.A. 85, 5946– Saenz, M. Pasquale-Styles, W. D. Dietrich, L. C. Weaver, The cellular
5950 (1988). Medline doi:10.1073/pnas.85.16.5946 inflammatory response in human spinal cords after injury. Brain 129, 3249–
17. R. J. Pasterkamp, R. J. Giger, M. J. Ruitenberg, A. J. Holtmaat, J. De Wit, F. De 3269 (2006). Medline doi:10.1093/brain/awl296
Winter, J. Verhaagen, Expression of the gene encoding the chemorepellent 35. H. Witte, D. Neukirchen, F. Bradke, Microtubule stabilization specifies initial
semaphorin III is induced in the fibroblast component of neural scar tissue neuronal polarization. J. Cell Biol. 180, 619–632 (2008). Medline
formed following injuries of adult but not neonatal CNS. Mol. Cell. Neurosci. 13, doi:10.1083/jcb.200707042
143–166 (1999). Medline doi:10.1006/mcne.1999.0738 36. M. Amit, M. K. Carpenter, M. S. Inokuma, C. P. Chiu, C. P. Harris, M. A. Waknitz, J.
18. K. C. Flynn, F. Hellal, D. Neukirchen, S. Jacob, S. Tahirovic, S. Dupraz, S. Stern, B. Itskovitz-Eldor, J. A. Thomson, Clonally derived human embryonic stem cell lines
K. Garvalov, C. Gurniak, A. E. Shaw, L. Meyn, R. Wedlich-Söldner, J. R. Bamburg, maintain pluripotency and proliferative potential for prolonged periods of
J. V. Small, W. Witke, F. Bradke, ADF/cofilin-mediated actin retrograde flow culture. Dev. Biol. 227, 271–278 (2000). Medline doi:10.1006/dbio.2000.9912
directs neurite formation in the developing brain. Neuron 76, 1091–1107 (2012). 37. Y. Shi, P. Kirwan, J. Smith, H. P. Robinson, F. J. Livesey, Human cerebral cortex
Medline doi:10.1016/j.neuron.2012.09.038 development from pluripotent stem cells to functional excitatory synapses. Nat.
19. D. G. Drubin, M. W. Kirschner, Tau protein function in living cells. J. Cell Biol. 103, Neurosci. 15, 477–486, S1 (2012). Medline doi:10.1038/nn.3041
2739–2746 (1986). Medline doi:10.1083/jcb.103.6.2739
20. R. Rouzier, R. Rajan, P. Wagner, K. R. Hess, D. L. Gold, J. Stec, M. Ayers, J. S. ACKNOWLEDGMENTS
Ross, P. Zhang, T. A. Buchholz, H. Kuerer, M. Green, B. Arun, G. N. Hortobagyi, Materials and methods and other supporting materials are available in the online
W. F. Symmans, L. Pusztai, Microtubule-associated protein tau: A marker of version of the paper. We thank L. Meyn, K. Weisheit, D. Fleischer and N. Thielen
paclitaxel sensitivity in breast cancer. Proc. Natl. Acad. Sci. U.S.A. 102, 8315– for technical assistance and animal care and Dr. C. Hill for teaching the spinal

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 3 / 10.1126/science.aaa2958


cord contusion injury model. We also thank C. Laskowski, C.H. Coles, A. Kania, M.
Hübener, W. Jackson and G. Tavosanis for critically reading and discussing the
manuscript. We are grateful for the support from the Human Spinal Cord Tissue
Bank and the electron microscopy core at the Miami Project as well as Professor
B. Kakulas, University of Western Australia and Royal Perth providing
anonymized post mortem sections following human spinal cord injury. This work
was supported by NIH, IRP, WfL and DFG. Harald Witte, Ali Ertürk, Farida Hellal,
and Frank Bradke filed a patent on the use of microtubule stabilizing compounds
for the treatment of lesions of CNS axons” (European Patent No. 1858498;
European patent application EP 11 00 9155.0; US patent application 11/908,118).
The authors declare no competing financial interests.
SUPPLEMENTARY MATERIALS
www.sciencemag.org/cgi/content/full/science.aaa2958/DC1
Materials and Methods
Figs. S1 to S11
Tables S1 and S2
Movies S1 to S8
References (30–37)

14 November 2014; accepted 25 February 2015


Published online 12 March 2015
10.1126/science.aaa2958

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 4 / 10.1126/science.aaa2958


Fig. 1. EpoB reduces inhibitory fibrotic scarring after SCI by abrogating meningeal fibroblast
polarization and migration. (A) Mass spectrometric analysis of CNS tissue and blood after single i.p.
injection of epoB, N = 4 rats/time-point. (B) Immunoblots (IB) of indicated proteins in lesion extracts, N
= 3 rats. (C) Human spinal cord after injury (asterisk), laminin immunolabeling. (D) Immunolabeling for
laminin, glial fibrillary acidic protein (GFAP) or chondroitin sulfates (CS-56) after rat spinal cord
hemisection. (E) Laminin-immunopositive (+) area at the lesion, N = 7 to 8 rats/group. (F)
Glycosaminoglycan amounts in spinal cord lesion extracts, N = 8 rats/group. (G) Rat meningeal
fibroblasts (RMFs) in wound healing assays. (H) Percentage of the area shown in (G) occupied with
RMFs after 48 hours, N = 3 experiments. (I and J) Immunolabeling of tyrosinated (TyrTub) and
detyrosinated tubulin (DetyrTub, arrowheads). (K) IB of indicated proteins in RMFs 24 hours after
treatment. (L) Immunolabeling for fibronectin, detyrosinated and tyrosinated tubulin (DAPI, nuclear
staining) in the rat meninges at the lesion. Bottom panel, magnification of fibroblasts (arrowheads) in
top panel. dpi, days post-injury. Scale bars, 50 μm. Schemes in (D) and (L) indicate lesion and displayed
region (red box). Values are plotted as means + SEM. *P < 0.05, ***P < 0.001 by Student’s t test.

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 5 / 10.1126/science.aaa2958


Fig. 2. EpoB promotes microtubule protrusion and axon elongation in neurons while
dampening microtubule dynamics in scar-forming fibroblasts. (A) Beta-3 tubulin (Tuj-1)
immunolabeling of neurons on inhibitory substrates (CSPGs, chondroitin sulfate
proteoglycans; Sema 3A, Semaphorin 3A). (B) Neurite length of cortical neurons after 48
hours under indicated conditions, N = 3-4 experiments. (C) EB3-mCherry time-lapse
projections in Nogo-A exposed neuron before and after epoB treatment (asterisks, stable
landmarks). Bottom panels, high magnification of boxed areas in top panels. (D) EB3-mCherry
fluorescence intensity in neurites under indicated conditions, N = 9-16 neurons (from 3
experiments). (E) Neurite growth on Nogo-A. Black arrowhead, time of indicated treatment. N
= 12-15 neurons (from 3 experiments). (F and G) EB3-mCherry time-lapse projections of
nocodazole treated neuron (F) and epoB treated meningeal fibroblast (G). Bottom panels, high
magnification of boxed areas in top panels. (H) EB3-mCherry time-lapse projections before
and after epoB treatment in cultured meningeal fibroblasts with (arrowhead) or without Tau-
expression. Bottom panels, magnification of boxed areas in top panels. (I) EB3-mCherry
fluorescence intensity in fibroblast periphery under indicated conditions, N = 20
cells/condition (from 4 experiments). Scale bars, 25 μm. Values are plotted as means (+ SEM
in (B) and (E)). *P < 0.05, **P < 0.01 by Student’s t test. n.s., not significant.

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 6 / 10.1126/science.aaa2958


Fig. 3. EpoB reduces dystrophy and promotes regeneration of injured spinal cord axons. (A)
Electron microscope images of human SCI. Top panel, undamaged axon containing microtubules
(black arrowheads). Bottom panel, retraction bulb (indicated by white arrowheads) without
microtubules. Middle panel, magnification of boxed area in bottom panel. Scale bars, 500 nm. (B)
Beta-3 tubulin (Tuj-1) immunolabeling of retraction bulbs in chronic human SCI. Scale bar, 10 μm.
(C) Lesioned GFP-positive spinal cord axons in mice forming retraction bulbs (yellow
arrowheads), dying back (red arrowheads) or regenerating (green arrowheads). Boxed area in top
panel, displayed region in panels below. Scale bars, 100 μm. (D and E) Percentage of injured
axons forming retraction bulbs (D) and distance between injured axons and injury site (E), N = 8
mice/group. Values are plotted as means + SEM. (F) Microruby-traced mouse dorsal column
axons after injury (white arrowheads), laminin and GFAP immunolabeling (dashed line, lesion
border). Scale bar, 100 μm. (G) Average distance between caudal lesion margin and injured axons
in individual animals (circles) and group means (vertical bars) ± SEM. *P < 0.05, **P < 0.01 by
Student’s t test.

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 7 / 10.1126/science.aaa2958


Fig. 4. EpoB promotes regrowth of raphespinal axons and improves walking after spinal
cord contusion injury. (A) Serotonin (5HT) immunolabeling (dashed line, lesion border) and
(B) number of 5HT-labeled (+) fibers caudal to a spinal dorsal hemisection, N = 7-8
rats/group. (C) Coronal sections of the lumbar spinal cord after contusion injury. Left panel,
co-immunostaining of 5HT, synaptophysin (Syn) and choline acetyltransferase (ChAT). Right
panels, magnification of each marker in boxed area (left panel) visualizing serotonergic
innervation of motor neurons (arrowheads). (D) Total length of 5HT-immunopositive fibers in
the ventral horn (5,7-DHT, 5,7-Dihydroxytryptamine), N = 4 (uninjured), 6 (7dpi), 11-12 rats
(56 and 70 dpi)/group. (E) Number of footfalls on the horizontal ladder, N = 10-11
rats/group. dpi, days post-injury. Scale bars, 50 μm. Schemes in (A) and (C) indicate lesion
and displayed region (red box). Values are plotted as means + SEM. *P < 0.05, n.s., not
significant by Student’s t test.

/ sciencemag.org/content/early/recent / 12 March 2015 / Page 8 / 10.1126/science.aaa2958

You might also like