bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1 Overexpression of Reticulon 3 enhances CNS regeneration and

2 functional recovery after injury

3

4 Sharif Alhajlah1,2, Adam M. Thompson1, Zubair Ahmed1*

5

6 1Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham,

7 Edgbaston, Birmingham, B15 2TT, UK.

8 2Applied Medical Science College, Shaqra University, Addawadmi, Riyadh, Saudi Arabia.

9

10

11 *Corresponding Author: Dr Zubair Ahmed, Neuroscience and Ophthalmology,

12 Institute of Inflammation and Ageing, College of Medical and Dental Sciences,

13 University of Birmingham, Birmingham, B15 2TT. Tel: +44 121 4148858; Fax: +44

14 121 4148867; email: [email protected].

15

16

17 ORCID ID: 0000-0001-6267-6442 (ZA).

18

19

20

21

22

23

24

25

26

1

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

27 Abstract

28 CNS neurons are generally incapable of regenerating their after injury due to

29 several intrinsic and extrinsic factors, including the presence of axon growth inhibitory

30 molecules. One such potent inhibitor of CNS axon regeneration is Reticulon (RTN) 4

31 or Nogo-66. Here, we focused on RTN3 as its contribution in CNS axon regeneration

32 is currently unknown. We found that RTN3 expression correlated with an axon

33 regenerative phenotype in dorsal root ganglion neurons (DRGN) after injury to the

34 dorsal columns, a model of . Overexpression of RTN3 promoted

35 disinhibited DRGN neurite outgrowth in vitro and dorsal column axon

36 regeneration/sprouting and electrophysiological, sensory and locomotor functional

37 recovery after injury in vivo. Knockdown of protrudin however, ablated RTN3-

38 enhanced neurite outgrowth/axon regeneration in vitro and in vivo. Moreover,

39 overexpression of RTN3 in a second model of CNS injury, the optic nerve crush

40 injury model, enhanced retinal ganglion (RGC) survival, disinhibited neurite

41 outgrowth in vitro and survival and axon regeneration in vivo, an effect that was also

42 dependent on protrudin. These results demonstrate that RTN3 enhances neurite

43 outgrowth/axon regeneration in a protrudin-dependent manner after both spinal cord

44 and optic nerve injury.

45

46

47

48

49 Key words: Reticulon-3, spinal cord injury, optic nerve injury, axon regeneration,

50 neurite outgrowth, protrudin.

2

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

51 Introduction

52 Reticulons (RTNs) are membrane-bound of which four have been identified

53 in and termed RTN1, 2, 3 and 4 (Nogo). RTNs are characterized by highly

54 conserved reticulon homology domains (RHD), approximately 150-200

55 that are located in RTN C-terminal, which are involved in improvement of localization

56 and function of these proteins. RTNs are found in a variety of tissues including the

57 brain, kidney, spleen and liver, playing diverse roles such as ER shaping and

58 morphology, membrane trafficking and . Moreover, RTNs are found in

59 neurodegenerative diseases such as Alzheimer’s, amyotrophic lateral sclerosis and

60 (Chiurchiu et al., 2014). RTN1, 2 and 4 have three splice isoforms

61 A, B and C whilst RTN3 only has two spliced isoforms RTN3A and B (Di Scala et al.,

62 2005). RTN3 is expressed in many types of tissues but was first isolated from

63 retina and later shown to have the highest expression in the brain(Cai et al., 2005).

64 The human RTN3 gene is localized in 11q13 and mutations of this gene cause

65 dysfunction of the retina and is therefore speculated that this gene may play a role in

66 retinal function (Kumamaru et al., 2004). Furthermore, RTN3 is also localised in

67 cultured cortical neurons and concentrated in axon growth cones in the early days

68 after culture (Kumamaru et al., 2004).

69

70 In neurons, a specific transport system for membrane-bound vesicles that contain

71 lipids and proteins is required for extension and maintenance of neurites (Futerman

72 and Banker, 1996; Tang, 2001). Kinesin-1 is a molecular that plays a key role

73 in this process, moving unidirectionally over long distances to mediate anterograde

74 transport of neuronal cargos from the cell body to the synapse (Ferreira et al., 1992;

75 Kimura et al., 2005; Hirokawa et al., 2009; Konishi and Setou, 2009). Kinesin-1

3

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

76 contains a dimer of kinesin heavy chain isoform 5A (KIF5) which hydrolyses

77 adenosine triphosphate (ATP) and uses the resulting energy to move along

78 microtubules (Hirokawa and Noda, 2008). Protrudin was identified as a key regulator

79 of Rab11-dependent vesicular transport during neurite extension through the

80 interaction of the atypical Rab11-binding domain with guanosine diphosphate (GDP)-

81 bound form of Rab11 (Shirane and Nakayama, 2006). Protrudin associated with

82 Rab11 in PC12 cells in response to protrudin phosphorylation, resulting from

83 signalling triggered by extracellular signal-related kinase (ERK) in response to nerve

84 growth factor (Shirane and Nakayama, 2006). In addition, expression of protrudin,

85 even in non-neuronal cells, induced the formation of membrane protrusions whilst

86 RNA interference (RNAi)-mediated downregulation of protrudin, inhibited neurite

87 formation (Shirane and Nakayama, 2006). Interestingly, protrudin associates with

88 KIF5 and its overexpression in HeLa cells induced the formation of protrusions

89 similar to those observed after protrudin overexpression (Matsuzaki et al., 2011).

90

91 Protrudin also interacts with RTN3 and promotes its binding to KIF5 allowing the

92 protrudin-KIF5 complex to cargo RTN3 from the cell soma to neurites (Matsuzaki et

93 al., 2011). More recently, protrudin was shown to form repeated contact sites with

94 late endosomes (LEs; the main of the secretory and endocytic pathways

95 along with the (ER)) via coincident detection of the small

96 GTPase RAB7 and phosphatidylinositol 3-phosphate (PtdIns(3)P), mediating transfer

97 of the microtubule kinesin 1 from protrudin to the motor adaptor FYCO1 (FYVE and

98 coiled-coil domain autophagy adaptor 1) on LEs (Raiborg et al., 2015). These

99 repeated LE-ER contacts promote microtubule-dependent translocation of LEs to the

100 cell periphery and subsequent synaptotagmin-VII (SYT7)-dependent fusion with the

4

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

101 plasma membrane, promoting neurite outgrowth (Raiborg et al., 2015). In this study,

102 we determined the contribution of RTN3 to CNS axon regeneration in a dorsal

103 column (DC) model of spinal cord injury (SCI) (Ahmed et al., 2014; Almutiri et al.,

104 2018; Farrukh et al., 2019; Stevens et al., 2019; Tuxworth, 2019). We then employed

105 a second model of CNS injury, optic nerve crush injury (Ahmed et al., 2006;

106 Tuxworth, 2019) to confirm our findings in the SCI model.

107

108

109 Results

110 Levels of RTN3 correlate with an axon regenerative phenotype

111 To examine changes in RTN3 mRNA levels, we harvested dorsal root ganglion

112 (DRG) containing the DRG neurons (DRGN) at 10 days after: (1), DC injury (non-

113 regenerating model); (2), sciatic nerve (SN) injury (regenerating SN model); (3), and

114 preconditioning (p) SN+DC lesion (crush of SN at -7 days before a DC lesion;

115 regenerating DC model) (Figure 1A). Preconditioning lesions upregulate the intrinsic

116 capacity of DRGN neurons to regenerate (Ahmed et al., 2010b; Farrukh et al., 2019).

117 We used microarray to demonstrate that RTN3 mRNA levels were significantly

118 elevated in SN and pSN+DC models, by 4.11 ± 0.02 and 6.22 ± 0.04-fold (P<0.0001,

119 ANOVA with Dunnett’s post hoc test; n = 16 DRG/group), respectively, compared to

120 intact controls (Figure 1B). These results were confirmed by qRT-PCR (Figure 1C)

121 and Western blot (Figure 1D,E) which demonstrated that regenerating SN and

122 pSN+DC regenerating models contained significantly elevated RTN3 levels. Low

123 levels of RTN3 immunoreactivity were detected in the and cell membranes

124 of DRGN after DC injury (Figure 1F). However, in regenerating SN and pSN+DC

125 DRGN, RTN3 immunoreactivity (green) was widely observed in the cytoplasm and

5

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

126 (Figure 1F). These results demonstrated that elevated levels of RTN3 correlated with

127 an axon regenerative phenotype and that RTN3 was mainly localised in the

128 cytoplasm of DRGN.

129

130 RTN3 is required for enhanced DRGN neurite outgrowth

131 To understand the contribution of RTN3 to DRGN neurite outgrowth, we

132 overexpressed RTN3 in freshly isolated primary adult DRGN using a non-viral vector,

133 in vivo-JetPEI (called PEI from herein), which we have shown produces similar

134 efficiencies of gene transduction as adeno-associated vectors (AAV8) in DRGN in

135 vitro and in vivo (Almutiri et al., 2018). Dissociated adult rat DRGN were allowed to

136 settle overnight, prior to treatment with plasmids containing control EGFP (non-

137 specific control) and RTN3, left to incubate in the presence of inhibitory

138 concentrations of CNS myelin extracts (CME), prepared and used at concentrations

139 as described by us previously (Ahmed et al., 2005) (Figure 2A). Western blot and

140 subsequent densitometry confirmed nearly 3-fold overexpression of RTN3 in treated

141 cultures, compared to EGFP-treated control cultures (Figure 2B,C). Overexpression

142 of RTN3 disinhibited DRGN neurite outgrowth such that DRGN were significantly

143 longer in length (Figure 2D,E) and the % of DRGN with neurites was also increased

144 (Figure 2F). However, overexpression of RTN3 did not affect DRGN survival (Figure

145 2G).

146

147 Since preconditioning lesions (pSN+DC) stimulate significantly higher levels of RTN3

148 mRNA and protein in DRGN, we dissociated DRGN from that had received a

149 preconditioning lesion at -7 days followed by a DC crush at day 0 and harvested

150 DRGN 10 days later (Figure 3A). We then used PEI to knock down RTN3 using an

6

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

151 shRNA plasmid DNA and grew them in the presence of neurite growth inhibitory

152 concentrations of CME, to determine DRGN survival and disinhibited neurite

153 outgrowth (Figure 3A). In a preliminary experiment, shRTN3 was titrated to determine

154 that 2µg of shRTN3 plasmids optimally caused 80% knockdown of RTN3 mRNA in

155 cultured DRGN (Figure 3 – figure supplement 1). Knockdown of RTN3 (with the

156 optimal amount of shRNA plasmid DNA) reduced RTN3 mRNA and protein by 80%

157 and 88%, respectively (Figure 3B-D). In DRGN cultures, knockdown of RTN3 almost

158 completely ablates DRGN neurite outgrowth without affecting survival (Figure 3E-H).

159 These results demonstrate that RTN3 is required for disinhibited DRGN neurite

160 outgrowth.

161

162 We then determined that overexpression of RTN3 using PEI in preconditioned DRGN

163 further upregulated RTN3 levels above that induced by preconditioning lesions alone

164 (Figure 4A-D). Overexpression of RTN3 in DRGN cultures from pSN+DC-treated

165 animals significantly potentiated DRGN neurite outgrowth in terms of both the mean

166 neurite length and % DRGN with neurites, over and above that observed in pSN+DC

167 cultures treated with a control EGFP vector (Figure 4E-G).

168

169 RTN3 overexpression has previously been shown to result in neurite abnormalities

170 and reduction in synaptic plasticity in transgenic mice by the aggregation of RTN3 in

171 RTN3 immunoreactive dystrophic neurites (RINDs) (Hu et al., 2007; Shi et al., 2013).

172 RINDs were abundantly present in a mouse model of Alzheimer’s disease (AD) and

173 hence RTN3 overexpression has been implicated in causing hippocampal age-

174 dependent RINDs. In our previous experiments, we did not observe any

175 abnormalities in DRGN neurite outgrowth. We tested whether overexpression of

7

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

176 RTN3 and subsequent exposure to Aβ1-42 resulted in DRGN abnormalities. However,

177 overexpression of RTN3 and exposure to Aβ1-42 had no significant effects on

178 disinhibited DRGN neurite outgrowth, DRGN survival nor were there any

179 abnormalities in DRGN neurite formation, such as dystrophic neurites (Figure 4 –

180 figure supplement 1). These results demonstrate that DRGN do not suffer adverse

181 effects of RTN3 overexpression as reported for hippocampal neurons.

182

183 Taken together, these results demonstrate that RTN3 overexpression promotes

184 DRGN neurite outgrowth in naïve DRGN cultures. DRGN exposed to preconditioning

185 lesions grow significantly longer neurites than naïve cultures and overexpression of

186 RTN3 in preconditioned DRGN further promotes disinhibited DRGN neurite

187 outgrowth. Since knockdown of RTN3 suppressed DRGN neurite outgrowth, this

188 suggests that DRGN require RTN3 for neurite outgrowth.

189

190 RTN3-mediated DRGN neurite outgrowth requires Protrudin signalling

191 We next sought to understand how RTN3 may signal to enhance DRGN neurite

192 outgrowth. Since the interaction of RTN3 with protrudin and protrudin to RAB7,

193 FYCO1 and SYST7 is important in regulating membrane protrusions resembling

194 neurites, we overexpressed RTN3 in preconditioned (pSN+DC injured) DRGN and

195 treated cultures with appropriate siRNAs to determine the contribution of protrudin,

196 FYCO1, RAB7 and SYST7 to disinhibited DRGN neurite outgrowth (Figure 5A).

197 Optimal concentrations of siRNAs to protrudin, RAB7, FYCO1 and SYST7 nearly

198 ablated all detectable levels of the relevant proteins in DRGN cultures (Figure 5B-E)

199 and completely suppressed DRGN neurite outgrowth observed after RTN3

200 overexpression (Figure 5F-K), without affecting DRGN survival (Figure 5L). These

8

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

201 results suggest that RTN3-mediated DRGN neurite outgrowth requires protrudin or

202 its binding partner signalling.

203

204 Since off-target effects of siRNA transfection such as activation of interferon

205 responses has been shown by us and others and appears to be siRNA sequence

206 dependent, we checked for a panel of interferon-mediated cytokines and innate

207 immunity-related related genes in our cultures after knockdown of protrudin, FYCO1,

208 RAB7 and SYT7 by qRT-PCR. We did not observe any activation of TNFα, IFNγ, IL-

209 6, IL-12, IL1β, MX1, IFIT, OAS1 or Casp7 mRNA in RTN3-overexpressed DRGN. In

210 contrast, transfection of RTN3-verexpressed DRGN cultures with a positive control

211 sip75NTR, which we know invokes a significant interferon response (Read et al.,

212 2009), induced 12-17-fold increases in mRNA of all of these genes (Figure 5 – figure

213 supplement 1).

214

215 Overexpression of RTN3 promotes DC axon regeneration and functional

216 recovery in vivo

217 To determine if RTN3 overexpression (Fig. S4A-C) promotes DC axon regeneration

218 in vivo and whether knockdown of Protrudin with shRNA (shProtrudin) ablates RTN3

219 overexpression-mediated axon regeneration over 6 weeks (Figure 6 – figure

220 supplement 1), we stained appropriately treated spinal cords with GAP43

221 to mark regenerating axons, since cholera toxin B labelling does not work in our

222 hands in the rat (Surey et al., 2014). Many GAP43+ axons were seen crossing the

223 lesion site (*) and reaching at least 6mm (>20%) into the distal segment of the spinal

224 cords of pSN+DC+PEI-RTN3-treated rats compared to little or no GAP43+ staining in

225 pSN+DC+PEI-EGFP-treated control spinal cords (Figure 6A,B). FluorRuby (FR)

9

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

226 anterograde tracer was used as a second method to confirm that GAP43+

227 regenerating axons were also co-labelled with FR in the same sections. However,

228 treatment with shProtrudin at the same time as RTN3 overexpression (pSN+DC+PEI-

229 RTN3+shProtrudin) ablated GAP43+ regenerating axons in spinal cords, normally

230 observed after RTN3 overexpression (pSN+DC+PEI-RTN3+shEGFP) (Figure 6A,B).

231

232 To determine if overexpression of RTN3 led to functional benefits, we used

233 electrophysiology, tape sensing and removal (sensory function) and ladder crossing

234 tests (locomotor function) (Almutiri et al., 2018; Stevens et al., 2019; Tuxworth,

235 2019). Individual CAP traces, CAP amplitudes and CAP areas that are normally

236 ablated after DC injury were significantly improved in RTN3 overexpressed animals

237 compared to controls (Figure 7A). However, RTN3-induced improvements in CAP

238 traces, amplitudes and areas, which were ablated when Protrudin was knocked down

239 (Figure 7A-C). Overexpression of RTN3 also promoted significant improvements in

240 sensory and locomotor function but again, these improvements were ablated if

241 Protrudin was also knocked down (Figure 7D,E).

242

243 These results demonstrate that overexpression of RTN3 promotes DC axon

244 regeneration after injury with return of significant beneficial sensory and locomotor

245 function.

246

247 Overexpression of RTN3 promotes upregulation of regeneration associated

248 genes after DC injury

249 We overexpressed RTN3 in preconditioned DRGN using PEI-delivered plasmids and

250 waited 10 days before harvesting DRG bundles and subjecting them to GAP43

10

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

251 immunostaining and qRT-PCR for regeneration associated genes (RAGs). We

252 observed that overexpression of RTN3 led to increased numbers of RTN3+ DRGN

253 (Figure 7 – figure supplement 1A), whilst qRT-PCR demonstrated significantly

254 increased expression of RAGs, namely gap43, sprr1a, cebp/epsilon, atf3 and galanin

255 (Figure 7 – figure supplement 1B). This suggests that RTN3 overexpression in

256 DRGN leads to activation of RAGs, which may account for the observed enhanced

257 axon regeneration and functional recovery seen after DC injury and RTN3

258 overexpression.

259

260 Overexpression of RTN3 promotes RGC survival and neurite outgrowth

261 We then employed a second CNS injury model to assess the contribution of RTN3 to

262 disinhibited neurite outgrowth/axon regeneration. We used the visual system, an

263 extension of the CNS where optic nerve crush (ONC) injury leads to rapid death of

264 retinal ganglion cells (RGCs) and axon regeneration can be promoted and easily

265 visualised (Ahmed et al., 2006; Yin et al., 2006; Fischer et al., 2008; Park et al.,

266 2008; Tuxworth, 2019). First, we used dissociated adult rat retinal cells, enriched in

267 RGCs, overexpressed RTN3 and cultured them for 3 days in the presence of

268 inhibitory concentrations of CME (Ahmed et al., 2006). We then performed Western

269 blot to confirm that RTN3 was overexpressed by >3-fold in cells treated with RTN3

270 plasmids than in EGFP-vector controls (Figure 8 – figure supplement 1A and B).

271 Staining for βIII-tubulin to mark RGC soma and neurites demonstrated that RTN3-

272 overexpression promoted significantly longer RGC neurites (Figure 8 – figure

273 supplement 1C and D) as well as increasing the % of RGC with neurites (Figure 8 –

274 figure supplement 1E), compared to controls (Neurobasal-A (NBA)+shEGFP) or the

275 positive control, ciliary neurotrophic factor (CNTF)) (Ahmed et al., 2006).

11

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

276 Interestingly, quantification of the number of βIII-tubulin+ RGC demonstrated that

277 overexpression of RTN3 was significantly more neuroprotective than the control

278 shEGFP plasmids, and as neuroprotective as the positive control, CNTF (Figure 8 –

279 figure supplement 1F). These results demonstrate that overexpression of RTN3 in

280 retinal cultures promotes RGC neurite outgrowth and survival.

281

282 Overexpression of RTN3 promotes RGC survival and axon regeneration after

283 optic nerve crush (ONC) injury

284 Finally, we assessed the contribution of RTN3 overexpression in RGC survival and

285 axon regeneration after optic nerve injury in vivo. Firstly, intravitreal injection of PEI-

286 RTN3 plasmids in vivo led to an almost 3-fold overexpression of RTN3 in the retina

287 (Figure 8A,B). Immunohistochemical staining with RTN3 antibodies demonstrated

288 localisation of RTN3, primarily to the ganglion cell layer (GCL) and inner plexiform

289 layer in ONC+shEGFP-treated control eyes, spreading to the inner (INL) and outer

290 nuclear layers (ONL) in ONC+RTN3-treated eyes (Figure 8C). High power images

291 demonstrated RTN3 (red) localisation to βIII-tubulin+ RGC (green; arrows) in the GCL

292 in both RTN3 and shEGFP vector control eyes (Figure 8D). RGC axon regeneration

293 in the optic nerve was significantly enhanced by overexpression of RTN3 compared

294 to shEGFP-vector control eyes, whilst co-expression of shProtrudin, attenuated

295 RTN3-induced RGC axon regeneration (Figure 8E,F). Like the in vitro results above,

296 overexpression of RTN3 also promoted significant RGC survival in vivo, 1294±145

297 and 1287±106 vs 447±25 in ONC+RTN3+shEGFP, ONC+RTN3+shProtrudin and

298 ONC+shEGFP, respectively (representing survival of 45% in ONC+RTN3+shEGFP-

299 treated eyes vs 20% in ONC+shEGFP-treated controls) with suppression of Protrudin

300 having no effect of RTN3-induced RGC survival (Figure 8G,H). Furthermore,

12

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

301 overexpression of RTN3 significantly improved photopic scotopic thresholds (pSTR),

302 a measure of RGC function, with protrudin suppression attenuating RTN3-induced

303 functional benefits (Figure 8I,J). These results demonstrate that overexpression of

304 RTN3 promotes RGC axon regeneration, survival and improved function after ONC.

305

306

307 Discussion

308 In this study, we show that RTN3 levels correlated with an axon regenerative

309 phenotype and that overexpression of RTN3 enhanced DRGN neurite outgrowth,

310 which was dependent on protrudin function. We also show that overexpression of

311 RTN3 in the DC model of SCI promotes significant DC axon regeneration, improved

312 electrophysiological, sensory and locomotor function. Furthermore, we used a

313 second model of CNS axon regeneration, the optic nerve crush (ONC) injury model,

314 to show that RTN3 is also required for enhanced RGC neurite outgrowth in vitro and

315 RGC axon regeneration in vivo, effects that were also dependent on protrudin.

316 Moreover, overexpression of RTN3 was also RGC neuroprotective in vitro and after

317 ONC in vivo.

318

319 It is increasingly clear that axon regeneration fails after CNS injury including SCI and

320 ONC due to a multitude of factors, including the low intrinsic capacity of CNS

321 neurons to regenerate their axons, the presence of a glial scar containing axon

322 growth inhibitory molecules and the presence of axon growth inhibitory molecules in

323 degenerating myelin (Mei et al., 2013; Kaplan et al., 2015; Fawcett and Verhaagen,

324 2018; Mahar and Cavalli, 2018). Some members of the reticulon family have also

325 been shown to inhibit CNS axon regeneration. For example, RTN4/Nogo-A is able to

13

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

326 bind to NgR1 and p75NTR and LINGO1/AMIGO3 to form a tripartite receptor complex

327 that transmits growth cone collapse signals into the cell via the Rho-ROCK pathway

328 (GrandPre et al., 2000; Ahmed et al., 2013). It was later shown that the

329 extracellular/ER luminal portion of Nogo-A, the 66-loop termed Nogo-66 (NgR1)

330 inhibited neurite outgrowth (GrandPre et al., 2000; Fournier et al., 2001). Genetic

331 ablation or pharmacological inhibition of the Nogo-A-NgR1 interactions promote axon

332 growth and behavioural recovery after SCI (GrandPre et al., 2002; Li et al., 2004; Cai

333 et al., 2005; Wang et al., 2006; Yang and Strittmatter, 2007). In addition, RTN1, 2

334 and 3 were reported not to inhibit axon regeneration (GrandPre et al., 2000).

335

336 However, we report here that upregulation of RTN3 correlated with both SN and

337 pSN+DC axon regeneration and its overexpression enhanced DRGN neurite

338 outgrowth/axon regeneration, leading to significant improvements in

339 electrophysiological, sensory and locomotor function after DC injury in rats. We

340 backed up these findings using a second CNS population of neurons, RGCs, which

341 also normally fail to regenerate their axons due to the same problems as is apparent

342 after SCI. However, significant RGC neurite outgrowth and axon regeneration was

343 promoted by overexpression of RTN3 in vitro and in vivo, effects that were also

344 protrudin dependent, suggesting a commonality between DRGN and RGC in their

345 response to RTN3. In contrast to the DC injury model, RTN3 overexpression was

346 neuroprotective for RGC both in vitro and after ONC in vivo, which was not the case

347 for DRGN where no effects of DRGN survival were observed. This is probably

348 explained by our observations that after ONC, significant RGC death occurs within 3

349 weeks after injury (Ahmed et al., 2005; Ahmed et al., 2010a; Ahmed et al.; Morgan-

350 Warren et al., 2016) whilst after DC injury, little or no DRGN death is observed

14

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

351 (Vigneswara et al., 2013). Hence, RTN3 overexpression in DRGN has little or no

352 protective effect as they rarely die in culture or after DC injury.

353

354 RTN3 overexpression and subsequent improvements in DRGN neurite outgrowth

355 were dependent on protrudin and its interacting molecules since knockdown of either

356 protrudin, RAB7, FYCO1 or SYT7 ablated RTN3-induced DRGN neurite outgrowth.

357 Moreover, RTN3-induced improvements in DC axon regeneration and

358 electrophysiological, sensory and locomotor function were also ablated in vivo by

359 suppression of protrudin. These results suggest that RTN3 works through protrudin

360 and its interacting molecules and that suppression of any of these ablates RTN3-

361 induced axon regeneration and subsequent functional improvements. Whether this is

362 a direct or indirect effect needs to be elucidated. However, protrudin normally forms

363 contact sites with late endosomes (LEs) via RAB7, mediating the transfer of kinesin-1

364 from protrudin to the motor adaptor FYCO1 on LEs. Repeated LE-ER contacts then

365 promote microtubule-dependent translocation of LEs to the cell periphery and

366 subsequent SYT-VII-dependent fusion with the plasma membrane, inducing neurite

367 outgrowth. Thus, it is possible that the interaction of RTN3 with protrudin is an

368 additional requirement for kinesin-1-bound protrudin-containing LE to be loaded onto

369 FYCO1 contact sites and translocation of LEs to the plasma membrane, promoting

370 protrusion and neurite/axon outgrowth. In addition, it is possible that RTN3

371 overexpression may enhance the handover of kinesin-1 that is bound to protrudin

372 containing LEs and hence movement of greater numbers of LEs to the plasma

373 membrane, enhancing neurite outgrowth/axon regeneration.

374

15

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

375 The improvements in sensory and locomotor recovery after RTN3 overexpression in

376 the spinal cord was remarkable since injured animals were indistinguishable from

377 sham-treated animals by 4 weeks after DC injury, suggesting significant recovery.

378 Even at 2 days after injury, there were significant improvements in sensory and

379 locomotor function, which we are unable to explain at present since it is likely to be

380 too soon for axonal regeneration/plasticity to account for these improvements. It is

381 possible that the improvements at 2 days after RTN3 overexpression may be due to

382 enhanced survival of injured cells in the spinal cord or greater preservation of cord

383 tissue architecture. This remains to be investigated.

384

385 In addition to promotion of axon regeneration and functional benefits in the DC injury,

386 we have shown that overexpression of RTN3 also promotes RGC survival, neurite

387 outgrowth, axon regeneration and restoration of significant RGC function by

388 electroretinography in ONC injury model. The ONC injury model is an excellent

389 model to evaluate neuroprotective and axon regenerative therapies since RGC soma

390 are easily accessible after intravitreal injection. Like our observations in the DC injury

391 model, RGC axon regeneration promoted by RTN3-overexpression was dependent

392 on protrudin function since knockdown of protrudin ablated RTN3-induced RGC

393 neurite outgrowth/axon regeneration and functional benefits. Moreover, RTN3 had an

394 RGC neuroprotective effect after ONC compared to DRGN after DC injury. This is

395 probably due to the fact that RGC death occurs rapidly after ONC whilst DRGN death

396 rarely occurs after DC injury. Hence, RTN3 probably exerts a neuroprotective effect

397 on both systems but is only evident after ONC due to the overt death of RGC that

398 normally occurs.

399

16

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

400 Conclusion

401 This study supports the notion that overexpression of RTN3 enhances CNS axon

402 regeneration, and in particular DC and RGC axon regeneration and thus leading to

403 improved sensory, locomotor and visual function. Our study therefore implies that

404 overexpression of RTN3 may be a possible treatment for SCI and ONC injury and

405 may restore lost function in affected patients.

406

407

408 Materials and Methods

409 Rats

410 All experiments were licenced by the UK Home Office and experimental protocols

411 were approved by the University of Birmingham’s Welfare and Ethical Review

412 Board (AWERB). All animal surgeries were carried out in strict accordance to the

413 guidelines of the UK Animals Scientific Procedures Act, 1986 and the Revised

414 European Directive 1010/63/EU. The ARRIVE guidelines were also followed in the

415 reporting of in vivo studies. Surgical procedures conformed to the guidelines and

416 recommendation of the use of animals by the Federation of the European Laboratory

417 Animal Science Associations (FELASA). Adult 6-8-week-old female Sprague-Dawley

418 rats weighing 170-220g (Charles River, Margate, UK) were housed in dedicated

419 facilities at the University of Birmingham. Up to four rats per cage were maintained in

420 a temperature-controlled environment on a 12-hour light/dark cycle with free access

421 to food and water ad libitum. All in vitro and in vivo experiments were performed with

422 the experimenter blinded to the treatment conditions and unblinded after data

423 analysis.

424

17

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

425 In vitro studies

426 Preparation of CNS myelin extracts (CME)

427 CME was prepared from adult rat brains as described by us previously (Ahmed et al.,

428 2005). Briefly, brains were homogenised in sucrose/EDTA and debris clarified by

429 centrifugation. The supernatant was layered on 0.9M sucrose and centrifuged at

430 20,000g for 60 min. The white material at the interface was collected and purified in

431 0.32M sucrose solution and the white pellet collected and freeze dried overnight. The

432 protein content of the CME was determined using the Pierce BCA assay (#5000001;

433 BioRad, Watford, UK) and the content of myelin-derived axon growth inhibitory

434 molecules such as myelin basic protein (#MCA409S; MBP; Serotec, Oxford, UK;

435 1:500 dilution), Nogo-A (#AB588; Sigma, Poole, UK; 1:500 dilution), myelin

436 associated glycoprotein (#MAB1567; MAG; Sigma; 1:500), and the CSPG brevican

437 (#SC-135849; Santa Cruz Biotechnology, CA, USA; 1:500 dilution) was determined

438 by western blot, as described below.

439

440 Rat primary DRGN cultures

441 Rat primary DRGN were prepared according to our previously published method

442 (Ahmed et al., 2005). Briefly, rats were killed by CO2 overexposure before harvesting

443 L4/L5 DRG pairs. Cells were dissociated into single cells using collagenase and

444 DRGN were grown in Neurobasal-A (NBA; #10888-022, Invitrogen, Paisley, UK) at a

445 plating density of 350 cells/well in 8-well chamber slides pre-coated with 100μg/ml

446 poly-D-lysine (#A-003-M, Sigma). CNS myelin extracts (CME) were prepared and

447 added to cultures at 100µg/ml to simulate the post-injury environment of the injured

448 spinal cord and incubated with or without appropriate treatments for 3 days at 37oC

449 and 5% CO2, as described previously (Ahmed et al., 2005). Experiments were

18

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

450 performed in triplicates and repeated on 3 independent occasions (n = 9

451 wells/condition).

452

453 Primary DRGN cultures after preconditioning SN lesion

454 Since preconditioning SN (pSN) lesioned DRGN contain significantly higher levels of

455 RTN3 mRNA/protein, we exposed the left SN at mid-thigh level and crushed the

456 nerve using fine forceps at the level of the sacrotuberous ligament. We then

457 harvested the ipsilateral L4/L5 DRGs at 7 days after injury dissociated them into

458 single cells and cultured them as described above.

459

460 Rat primary retinal cell cultures

461 Retinal cells were dissociated into single cells using a Papain dissociation kit

462 (#LK003150, Worthington Biochemicals, Lakewood, NJ, USA), as described by us

463 previously (Ahmed et al., 2006). Briefly, 125 x 103 cells were plated in 8-well

464 chamber slides in NBA pre-coated with 100μg/ml poly-D-lysine and CME and

o 465 incubated with or without appropriate treatments for 3 days at 37 C and 5% CO2

466 (Ahmed et al., 2006). Experiments were performed in triplicates and repeated on 3

467 independent occasions (n = 9 wells/condition).

468

469 DRGN/RGC neurite outgrowth and survival

470 DRGN or retinal cells were fixed in 4% PFA, washed in several changes of PBS and

471 immunocytochemistry performed as described previously (Ahmed et al., 2005;

472 Ahmed et al., 2006). Briefly, DRGN/RGC were stained with βIII-tubulin antibodies

473 (#T8578 Sigma; diluted 1:200) to label the DRGN/RGC somata and neurites. With an

474 investigator masked to the treatment conditions, each well of a chamber slide was

19

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

475 split into 9 quadrants and images were captured randomly using an Axioplan 200 epi-

476 fluorescent microscope, equipped with an Axiocam HRc and Axiovision Software (all

477 from Carl Zeiss, Hertfordshire, UK). Axiovision was then used to measure the length

478 of the longest DRGN neurite from at least 30 DRGN/well and all RGC/well. The

479 number of DRGN/RGC with neurites and the total number of surviving DRGN were

480 also recorded as described previously (Ahmed et al., 2005; Ahmed et al., 2006).

481 Overexpression of RTN3 in DRGN and retinal cultures

482 To overexpress RTN3 in DRGN and retinal cells in vitro, a CMV promoter driven rat

483 RTN3 plasmid was purchased from Origene, Rockville, MD, USA (#RR200211L3).

484 Transfection controls were carried out using a CMV driven, non-targeting, EGFP

485 plasmid (#PS100065; Origene). In preliminary experiments, optimal concentrations of

486 RTN3 plasmids were determined as 2µg. Lipofectamine 2000 (#11668027; LF2000;

487 Invitrogen) was prepared according to the manufacturer’s instructions and used to

488 deliver 2µg of RTN3 (PEI-RTN3) or EGFP (PEI-EGFP) plasmids were used to

489 transfects DRGN and retinal cells. Additional controls included untreated

490 DRGN/retinal cells or PEI alone. DRGN and retinal cells were allowed to incubate for

491 3 days prior to harvesting of cells and extraction of total protein/RNA for validation of

492 RTN3 protein/mRNA overexpression.

493

494 Knockdown of RTN3, protrudin, FYCO1, RAB7 and SYT7 using siRNA

495 For siRNA transfection, a preliminary dose determination experiment was run and

496 5nM siRTN3 delivered using LF2000 (Invitrogen) was used to determine optimal

497 knockdown of RTN3 mRNA/protein in DRGN/RGC cultures, as described previously

498 (Morgan-Warren et al., 2016). The efficiency of knockdown was assessed using

499 quantitative RT-PCR (qRT-PCR) or western blot, as described below. RTN3 was

20

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

500 knocked down in DRGN/retinal cultures using an siRNA to rat RTN3 (siRTN3; Assay

501 ID 251374, cat no. AM16708, ThermoFisher Scientific). Protrudin (siProtrudin; Assay

502 ID 196855, cat no. AM16708), FYCO1 (siFYCO1; Assay ID, s236101, cat no.

503 4390771, ThermoFisher Scientific), Rab7 (siRAB7; Assay ID 52712, cat no.

504 AM16708, ThermoFisher Scientific), SYT7 (siSYT7; Assay ID 199167, cat no.

505 AM16708, ThermoFisher Scientific) was used to knock down appropriate targets in

506 DRGN/retinal cultures, delivered using LF2000. An siRNA to EGFP was also used as

507 a non-targeted control for transfections (Morgan-Warren et al., 2016). Experiments

508 were performed in triplicate and repeated on 3 independent occasions (n = 9

509 wells/condition).

510

511 Quantitative RT-PCR (qRT-PCR) in DRGN and retinal cultures

512 DRGN and retinal cells were washed with PBS and total RNA (n = 6-9

513 wells/treatment) was extracted using Trizol reagent according to the manufacturer’s

514 instructions (Invitrogen). Appropriate primers were used to validate each gene of

515 interest using the specific primers (Table 1; (Almutiri et al., 2018)) or pre-validated

516 primer sequences (Table 2). qRT-PCR was performed on a LighCycler Q-PCR

517 machine (Roche, Burgess Hill, UK) following published methods (Read et al., 2009).

518 Fold changes were computed using ΔΔCt method.

519 520 Tissue/cell lysis, western blot and densitometry

521 Total protein was extracted from L4/L5 DRG, DRGN cultures, retinal cells and retinae

522 after lysis in ice cold lysis buffer as appropriate and subjected to Western blot as

523 described previously (Ahmed et al., 2005; Ahmed et al., 2006). A DC protein assay

524 kit (#5000111, Bio-Rad, Watford, UK) was then used to determine total protein

525 content and 40µg of total protein from each relevant sample was separated on 12%

21

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

526 SDS-PAGE, transferred onto PVDF membranes (ThermoFisher Scientific #88520)

527 and probed for: RTN3 (rabbit polyclonal anti-RTN3; Merck Millipore #ABN1723,

528 Watford, UK; diluted 1:500); Protrudin (Rabbit anti-Protrudin; MyBiosource

529 #MBS8570624; diluted 1:500); FYCO1 (Rabbit anti- FYCO1; MyBiosource

530 #MBS9600315, San Diego, CA, USA; diluted 1:500); RAB7 (Rabbit anti-RAB7;

531 Abcam #ab137029, Cambridge, UK; diluted 1:500); and SYT7 (Mouse anti-SYT7;

532 MYBioscource #MBS800258) in 5% non-fat milk. Bands were detected using HRP-

533 labelled anti-rabbit IgG and developed using an enhanced luminescence kit (GE

534 Healthcare #GENA934, Watford, UK). Beta actin (Sigma #A5316, Poole, UK; 1:1000)

535 was used as a loading control. An investigator masked to the treatment conditions

536 quantified bands from 3 repeats by densitometry using the built-in-gel plotting macros

537 in Image J (https://imagej.nih.gov/ij/index.html), as described previously (Ahmed et

538 al., 2009).

539

540 In vivo studies

541 Experimental Design

542 Animals were randomly assigned to treatment groups and experimenters were

543 masked to the treatment conditions. Sample sizes were no computed using power

544 calculations, instead these are based on previous similar experiments published by

545 us (Almutiri et al., 2018; Tuxworth et al., 2019; Kitchen et al., 2020) Unless otherwise

546 stated, all in vivo experiments were performed with n = 4 rats/group/test and each

547 experiment repeated on at least 2-3 independent occasions (total n = 8-12

548 rats/group), except for the electrophysiology, ladder crossing and tape

549 sensing/removal tests where n = 6 rats/group were used and repeated on 3

22

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

550 independent occasions (total n = 18 rats/group). The same rats were used for

551 electrophysiology, ladder crossing and tape sensing/removal tests.

552

553 For the in vivo microarray experiments, 5 groups of animals each comprised of 4

554 adult female Sprague-Dawley rats were designated as: (1), uninjured intact controls

555 (IC); (2) sham treated controls (Sham: partial laminectomy but no DC crush injury)

556 (3), non-regenerating DC model (DC) that received a DC lesion alone; (4),

557 regenerating SN model (SN) that received an SN lesion alone; and (5), regenerating

558 pre-conditioning (p) SN lesion (pSN)+DC injury model (pSN+DC - pSN lesion

559 performed 7 day (d) before the DC lesion). The ipsilateral L4/L5 DRG were harvested

560 from each animal at 10d after DC, SN and pSN+DC lesions, pooled together and

561 RNA extracted for microarray (n = 8 DRG/group (4 rats/group)), qRT-PCR (n = 16

562 DRG/group (4 rats/group, 2 independent repeats)) and immunohistochemistry

563 studies (n = 16 DRG/group (4 rats/group, 2 independent repeats)).

564

565 To stimulate the intrinsic capacity of DRGN to grow neurites in vivo (‘regeneration

566 readiness’), rats received a unilateral pSN lesion 7d prior to harvesting of affected

567 L4/L5 DRG (Neumann and Woolf, 1999; Ahmed et al., 2010b). In vivo stimulated

568 DRGN were then dissociated into single cells using collagenase, cultured and treated

569 as described in the appropriate section below.

570

571 RTN3 overexpression

572 To overexpress RTN3 in DRGN or retina in vivo, a CMV promoter driven rat RTN3

573 plasmid was purchased from Origene, Rockville, MD, USA (Cat no. RR200211L3). A

574 CMV driven, non-targeting, EGFP plasmid (Cat No. PS100065) was used as control.

23

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

575 Plasmid DNA for either RTN3 or the control EGFP vector were delivered by intra-

576 DRG injection into L4/5 DRG or intravitreal injection into the eye, respectively,

577 condensed in in vivo-JetPEI (PEI; #201, Polyplus Transfection, New York, USA), a

578 non-viral vector that we have shown to be as efficient at transducing neurons as

579 efficiently as AAV8 (Jacques et al., 2012; Almutiri et al., 2018). PEI was prepared

580 according to the manufacturer’s instructions and in preliminary experiments, 2µg of

581 plasmid DNA was determined to be optimal for RTN3 overexpression in vitro and in

582 vivo (not shown). An equivalent amount of the GFP plasmid was used to control for

583 RTN3 transfection. In further experiments pre-optimised concentrations of plasmids

584 were then used to monitor the time-frame of RTN3 overexpression in DRGN and

585 retina in vivo at 72 hours, 1 week, 2 weeks, 4 weeks and 6 weeks by qRT-PCR, and

586 at 14 days by western blot and immunohistochemistry after intra-DRG or intravitreal

587 injection. PEI maintains overexpression for at least 6 weeks in both DRGN and

588 retina.

589

590 To test the potential of RTN3 to promote DC axon regeneration and improve

591 functional recovery, groups of 6 adult Sprague-Dawley rats (total n = 18 rats/group

592 from 3 independent repeats) received pSN lesions 7d before DC lesion to stimulate

593 regeneration ‘readiness’. Immediately after DC injury at day 0, animals received intra-

594 DRG injection of in vivo-jetPEI (referred to as PEI from herein), PEI-GFP or PEI-

595 RTN3 and allowed to recover for 6 weeks during which behavioural tests were

596 performed at baseline, 2 days and then weekly at 1-6 weeks after injury (Fagoe et al.,

597 2016; Almutiri et al., 2018). Electrophysiology to record compound action potentials

598 (CAP) were also performed on the same set of animals at 6 weeks after injury before

599 harvesting tissues for immunohistochemistry and determination of axon regeneration

24

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

600 by GAP43 (#33-5000; mouse monoclonal anti-GAP43 ; Invitrogen, 1:400

601 dilution) immunohistochemistry (Almutiri et al., 2018).

602

603 Some animals (n = 6/group) also received 0.5 µl of the bi-directional tracer,

604 FluoroRuby ((5% in saline) MW 10,000; ThermoFisher Scientific), injected slowly

605 using a micropipette with a tip diameter between 30-40µm, at a depth of 0.5mm and

606 10mm caudal to the lesion site 1 day before animal sacrifice. Rats were then

607 intracardially perfused with 4% paraformaldehyde, cryoprotected in sucrose,

608 embedded in optimal cutting temperature (OCT; Raymond A Lamb, Peterborough,

609 UK) embedding medium and longitudinal sections of the spinal cord cut at a

610 thickness of 15µm using a cryostat, as described in the immunohistochemistry

611 section below.

612

613 For RGC axon regeneration, groups of 4 adult Sprague-Dawley rats (3 independent

614 repeats; total n = 12 rats/group) received intravitreal injection of RTN3 or the control

615 EGFP vector and were allowed to recover for 28d after optic nerve crush injury

616 (ONC). Optic nerve regeneration was then assessed using the gold standard method

617 in the rat of GAP43 immunohistochemistry, as described later.

618

619 Protrudin shRNA in vivo

620 DNA fragments encoding stem loop–type shRNAs specific for human protrudin

621 mRNA (5’-GCTGAGGTGAAGAGCTTCTTG-3’) or GFP (non-specific control) mRNA

622 (5′ GCTGACCCTGAAGTTCATC-3′) were synthesised, attached to the U6 promoter

623 and subcloned into pBluescriptII SK(+) vector (Stratagene #212205, La Jolla, CA,

624 USA), as described previously (Matsuzaki et al., 2011). The efficiency of knockdown

25

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

625 was assessed by introduction of various concentrations of the plasmid vectors using

626 in vivo-JetPEI by intra-DRG (L4/5) or intravitreal injection in vivo, as described above.

627 In preliminary experiments, the optimal concentration of plasmid DNA to knock down

628 Protrudin in vivo was determined as 2µg (not shown). Knockdown efficiency with

629 preoptimized concentrations of plasmids was assessed at 72 hours, 1 week, 2

630 weeks, 4 weeks and 6 weeks (n = 4 DRG/time-point (2 rats/time-point), 3

631 independent repeats (total 12 DRG/time-point (6 rats/time-point) or 6

632 retinae/timepoint), by qRT-PCR analysis, with cells expressing control non-targeting

633 EGFP shRNA.

634

635

636 Dorsal column crush injury

637 For DC and SN crush lesions, rats received subcutaneous Buprenorphine injections

638 before anaesthesia (5% isoflurane with 1.8l/min O2). After a partial laminectomy, DC

639 were crushed bilaterally at the level of T8 using calibrated watchmaker’s forceps

640 (Lagord et al., 2002; Ahmed et al., 2010c; Surey et al., 2014). The tips of the forceps,

641 separated to a width of 1mm, were inserted into the cord through the dorsal

642 meninges to a depth of 1mm and the DC crushed by tip approximation. For SN or

643 pSN lesion, the left SN was exposed at mid-thigh level and crushed using fine

644 forceps at the level of the sacrotuberous ligament. In the pSN+DC model, SN were

645 crushed 1 week before DC crush injury. Pre- and post-surgical analgesia was used

646 as standard and as advised by the Named Veterinary Surgeon.

647

648 FluoroRuby Tracer injection

26

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

649 Since Cholera toxin B labelling does not work in the rat in our hands (Ahmed et al.,

650 2014), we used FluoroRuby bidirectional tracer to confirm regenerating axons in the

651 DC. A micropipette with a tip diameter between 30-40µm was used to inject 0.5 µl of

652 5% FluoroRuby (MW 10,000; ThermoFisher Scientific, Leicester, UK) dissolved in

653 0.9% saline at a depth of 0.5mm, 10mm caudal to the lesion site 1 day before animal

654 sacrifice. Rats were then intracardially perfused with 4% paraformaldehyde,

655 cryoprotected in a graded series of sucrose, embedded in optimal cutting

656 temperature (OCT; Raymond A Lamb, Peterborough, UK) embedding medium and

657 longitudinal sections of the spinal cord cut at a thickness of 15µm using a cryostat, as

658 described in the immunohistochemistry section below. The same sections were

659 stained for GAP43+ and FR to demonstrate co-staining, as described below.

660

661 Optic nerve crush (ONC) injury

662 After injection of rats with Buprenorphine to provide analgesia, rats were

663 anesthetised using 5% isoflurane in 1.8ml/l of O2 with constant body temperature and

664 heart rate monitoring. Optic nerves were unilaterally crushed 2mm from the lamina

665 cribrosa using a calibrated watchmaker’s forceps, as described by us previously

666 (Ahmed et al., 2005). In general n = 4 rats/group were used and each experiment

667 was repeated on 3 independent occasions (total n = 12 rats/group).

668

669 FluroGold back-labelling of RGC

670 FluorGold (FG) retrograde tracer was used to count the number of surviving RGC in

671 retinal wholemounts as described by us previously (Ahmed et al., 2011). Briefly, 2

672 days before killing rats, 4% FluroGold (FG; Cambridge Bioscience, Cambridge, UK)

673 retrograde tracer was injected into the nerve mid-way between the lamina cribrosa

27

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

674 and the site of ONC. After 48 hours, animals were killed by CO2 overexposure and

675 animals were intracardially perfused with 4% formaldehyde, retinae dissected out,

676 immersion fixed and whole mounted onto glass slides. Images were captured from

677 three different areas of each retinal quadrant (total n = 12 counts per quadrant, n = 4

678 retinas per treatment, to account for variation of RGC numbers in the different areas

679 and quantified using the built-in particle counting facilities in ImagePro; Media

680 Cybernetics, Bethesda, MD, USA) and expressed as the number of RGC per mm2 ±

681 S.E.M.

682

683 Tissue harvesting and processing

684 For the microarray and western blot studies, rats were killed at 10 days after DC

685 lesion by CO2 overexposure and freshly isolated L4/L5 DRG, spinal cord, retinae or

686 optic nerves from appropriate experiments were harvested and snap frozen in liquid

687 N2 before RNA extraction for quantitative RT-PCR (qRT-PCR) or protein extraction

688 for Western blot, as described later.

689

690 For immunohistochemistry, animals were intracardially perfused with 4%

691 paraformaldehyde (PFA; #F007, TAAB Laboratories, Peterborough, UK) in

692 phosphate buffered saline (PBS; #P4417, Sigma, Poole, UK) and both ipsilateral and

693 contralateral L4/L5 DRG and lesion site plus 5mm either side of the spinal cord and

694 retinae/optic nerves were dissected out and post-fixed for 2 hours at 4oC. All tissues

695 were then cryoprotected in a graded series of sucrose, blocked up in optimal cutting

696 temperature embedding compound (#F320/B1, TAAB laboratories) and sectioned

697 through a para-sagittal plane on a cryostat set at 15µm-thick. Sections from the entire

28

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

698 DRG, spinal cord, retinae or optic nerves were collected onto charged slides and

699 stored at -20oC until required, noting the middle sections of each sample.

700

701 Microarray analysis after DC injury

702 RNA extraction and microarray analysis was performed as described

703 previously(Read et al., 2009). Briefly, total RNA was extracted, hybridised with Cy3-

704 or Cy5-labelled oligonucleotide probes and the eventual slides scanned with Axon

705 GenePix 4000B scanner (Molecular Devices Ltd, Berkshire, UK). Data were analysed

706 using GeneSpring GX7 (Agilent, Berkshire, UK), normalised by the Lowess method

707 and filtered between the P<0.05 threshold and fold-changes >2 above sham control

708 levels were deemed significant (Read et al., 2009).

709

710

711

712 Quantitative RT-PCR (qRT-PCR)

713 The levels of RTN3 mRNA were validated by qRT-PCR using a pre-validated primer

714 sequence (rat RTN3, Rn01498010_m1; cat no. 4351372; ThermoFisher Scientific,

715 Loughborough, UK). Levels of protrudin mRNA were analysed using pre-validated

716 primer sequences (rat Protrudin, RN01766356_m1; cat no. 4331182; ThermoFisher

717 Scientific). Q-PCR was performed on a LighCycler Q-PCR machine (Roche, Burgess

718 Hill, UK) following published methods (Read et al., 2009). Fold changes were

719 computed using ΔΔCt method.

720

721 Immunohistochemistry

29

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

722 The middle sections of each DRG, spinal cord, optic nerve or retinae were chosen for

723 immunohistochemistry as described by us previously (Ahmed et al., 2011; Surey et

724 al., 2014; Vigneswara et al., 2014; Almutiri et al., 2018). One-three sections per

725 biological replicates was analysed and quantified for each group, and the number of

726 biological replicates per group is indicated in the figure legends. Briefly, masked

727 slides were allowed to thaw at room temperature before being washed in PBS. Non-

728 specific antibody binding was then blocked with 3% bovine serum albumin diluted in

729 PBS containing 0.1% Triton X-100 before incubation with relevant antibody overnight

730 at 4oC in a humidified chamber. DRG were co-stained with rabbit polyclonal anti-

731 RTN3 (#ABN1723, Merck Millipore; 1:400 dilution) and monoclonal anti-NF200

732 (#N5389, Sigma, 1:400) primary antibodies. RGC in the retina were co-stained with

733 RTN3 as above and monoclonal anti-βIII-tubulin (#T8578, Sigma; 1:200). GAP43

734 was used to detect regenerating axons in the cord/optic nerve and detected with

735 mouse anti-GAP43 (#33-5000 (clone 7B10), Invitrogen; 1:400) primary antibody.

736 GAP43 was used since Cholera toxin B labelling in our hands did not label

737 regenerating DC axons in the rat (Surey et al., 2014). After several washes in PBS,

738 Alexa488 anti-rabbit (#A323731, Invitrogen)/Texas Red anti-mouse (#PA1-28626,

739 Invitrogen) (RTN3 and NF200+/βIII-tubulin staining) and Alexa488 anti-mouse

740 (#A32723, Invitrogen) (to detect GAP43+ staining) were diluted 1:400 in PBS and

741 incubated for 1hr at room temperature in a humidified chamber. Sections were then

742 washed in several changes of PBS and coverslips mounted using Vectashield

743 containing DAPI (#H-1900, Vector Laboratories, Peterborough, UK). Negative

744 controls, including omission of primary antibody, were included in each run and were

745 used to set the background threshold level for each antibody before image capture.

746 Slides were viewed using an Axioplan 200 epi-fluorescent microscope equipped with

30

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

747 an Axiocam HRc and run using Axiovision Software (all from Zeiss, Herefordshire,

748 UK)

749

750 Other antibodies for immunocytochemistry/immunohistochemistry

751 The following antibodies were used for immunoassays: primary antibodies - rabbit

752 polyclonal anti-RTN3 (#ABN1723, Merck Millipore; 1:400 dilution), monoclonal anti-

753 NF200 (#N5389, Sigma, 1:400), monoclonal anti-βIII-tubulin (#T8578, Sigma; 1:200),

754 mouse anti-GAP43 (#33-5000 (clone 7B10), Invitrogen; 1:400); secondary antibodies

755 - Alexa488 anti-rabbit (#A323731, Invitrogen; 1:400 dilution), Texas Red anti-mouse

756 (#PA1-28626, Invitrogen; 1:400 dilution), Alexa488 anti-mouse (#A32723, Invitrogen;

757 1:400 dilution).

758

759 Quantification of GAP43+ axons

760 Regenerating GAP43+ axons in the spinal cord were quantified by collecting all serial

761 parasagittal 50um-thick sections (70-80 sections/rat; n = 6 rats/group, 3 independent

762 repeats; total n = 18 rats/treatment) and counting the number of GAP43+ fibres

763 through a dorsoventral oriented line from 4mm rostral to 4 mm caudal to the lesion

764 site, as described previously (Hata et al., 2006; Almutiri et al., 2018). Axon number

765 was calculated as a percentage of the number of GAP43+ fibres seen 4mm above

766 the lesion site where the DC was intact.

767

768 Regenerating GAP43+ axons in the optic nerve were quantified from 3 sections/optic

769 nerve (n = 4 optic nerves/group, 3 independent repeats; total n = 12 optic

770 nerves/group) at 0.2, 0.5, 1, 1.5, 2, 2.5, 3 and 4mm from the lesion epicentre. GAP43

771 in the optic nerve correlates with the number of axons detected by the anterograde

31

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

772 tracer, Rhodamine B, and thus accurately demonstrates regenerating RGC axons

773 (Vigneswara et al., 2014). The cross-sectional width of the optic nerve was also

774 recorded where axon counts were performed and used to calculate the number of

775 axons/mm optic nerve width. The total number of axons extending distance d in an

776 optic nerve with a radius r, Σad, was calculated using the formula:

2 777 Σad = πr x (average number of axons/mm width)/(section thickness, t, 0.015mm)

778

779 Electroretinography

780 Electroretinography (ERG) was performed as described by us previously (Mead et

781 al., 2020). Briefly, rats were dark adapted for 12 hours and after anaesthesia eyes

782 were dilated with tropicamide. Scotopic flash ERG was recorded under dim red light

783 from -5.5 log (cd s) m-2 to 1.0 log (cd s) m-2 in o.5 log unit increments. Traces were

784 analysed using the built-in Espion software and traces at a light intensity of -5.5 log

785 (cd s) m-2 were chose for analysis as they produced clean, unambiguous positive

786 scotopic thresholds (pSTR) at 100 ms after stimulus. Peak amplitude of the pSTR

787 was recorded by an experimenter masked to the treatment conditions.

788 Electrophysiology

789 At 6 weeks after surgery or treatment, compound action potentials (CAP) were

790 recorded after vehicle, RTN3 overexpression or RTN3 overexpression/Protrudin

791 suppression (n = 6 rats/group, 3 independent repeats (total n = 18/group/test)) as

792 previously described (Lo et al., 2003; Hains et al., 2004; Almutiri et al., 2018). Briefly,

793 with the experimenter masked to the treatment conditions, silver wire electrodes were

794 used to apply single-current pulses (0.05ms) through a stimulus isolation unit in

795 increments (0.2, 0.3, 0.6, 0.8, and 1.2 mA) at L1-L2 and compound action potentials

796 (CAP) recorded at C4-C5 along the surface of the midline spinal cord. CAP

32

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

797 amplitudes were calculated between the negative deflection after the stimulus

798 artefact and the next peak of the wave. CAP area was calculated by rectifying the

799 negative component (full-wave rectification in Spike 2 software) and measuring its

800 area at the different stimulation intensities. At the end of the experiment, the dorsal

801 half of the spinal cord was transected between stimulating and recording electrodes

802 to confirm that a CAP could not be detected. Spike 2 software (Cambridge Electronic

803 Design, Cambridge, UK) was used to analyse the data and representative Spike 2

804 software processed CAP traces data are shown.

805

806 Functional tests after DC injury

807 Functional testing was carried out as described by us previously (Fagoe et al., 2016;

808 Almutiri et al., 2018; Tuxworth, 2019; Kitchen et al., 2020). Briefly, animals (n = 6

809 rats/group, 3 independent repeats (total n = 18/group/test)) received training to

810 master traversing the horizontal ladder functional testing. Baseline parameters for all

811 functional tests were established 2-3 days before injury. Animals were then randomly

812 assigned and treatment status masked from the investigators and tested at 2 day

813 after DC lesion + treatment and then weekly for 6 weeks. Experiments were

814 performed by 2 observers masked to the treatment status, with animals being tested

815 in the same order and at the same time of day. Three individual trials were performed

816 each time for each animal for each test.

817 Horizontal ladder crossing test. This tests the animals’ locomotor function and is

818 performed on a 0.9-meter-long horizontal ladder with a diameter of 15.5 cm and

819 randomly adjusted rungs with variable gaps of 3.5-5.0 cm. The total number of steps

820 taken to cross the ladder and the number of left rear paw slips were recorded and the

33

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

821 mean error rate was then calculated by dividing the number of slips by the total

822 number of steps taken.

823

824 Tape removal test (sensory function). The tape removal test determines touch

825 perception from the left hind paw. Animals were held with both hind-paws extended

826 and the time it took for the animal to detect and remove a 15x15mm piece of tape

827 (Kip Hochkrepp #803, Bocholt, Germany) was recorded and used to calculate the

828 mean sensing time.

829

830 Statistical analysis and data visualisation

831 Data are presented as mean ± SEM. Normally distributed data were interrogated by

832 one-way analysis of variance (ANOVA), with Dunnett’s post hoc tests, set at P<0.05

833 in SPSS (IBM, NJ, USA). All P and n values are specified within each figure legend.

834 No statistical tests were used to predetermine sample sizes, but our sample sizes

835 were similar to those reported in previous publications (Ahmed et al., 2014; Almutiri

836 et al., 2018; Farrukh et al., 2019; Stevens et al., 2019; Tuxworth, 2019). All images

837 shown are representative images of data quantified in corresponding graphs.

838 For the horizontal ladder test, data were analysed by comparing lesion versus sham-

839 treated rats, using binomial generalised linear mixed models (GLMM), with

840 lesioned/sham ('LESION'; set to true in lesioned animals post-surgery, false

841 otherwise) and operated/unoperated ('OPERATED'; set to false before surgery, true

842 after surgery) as fixed factors, animals as a random factors and time as a continuous

843 covariate (Almutiri et al., 2018; Tuxworth, 2019; Kitchen et al., 2020). Data were fitted

844 with lme4 with the glmer function and P values calculated using parametric bootstrap

845 with pkbrtest in R package (www.r-project.org), set to between 1000 and 20000

34

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

846 simulations, as described previously (Fagoe et al., 2016; Almutiri et al., 2018). For

847 the tape removal test, linear mixed models (LMM) were compared with lme4 and

848 glmer functions in R package. P values were then calculated in R using pkbrtest, with

849 the Kenward-Roger method (Fagoe et al., 2016; Almutiri et al., 2018).

850

851 Data availability

852 All data generated during this study are included in the manuscript. Source files are

853 available.

854

855

856 Acknowledgements

857 The authors would like to thank the staff at the University of Birmingham’s Biomedical

858 Services Unit for their technical assistance with animal work described in this

859 manuscript, including randomisation and masking treatment groups.

860

861

862

863 Funding

864 This work was supported by the Saudi Education Ministry grant SHU11 (S.A., and

865 Z.A.), and funding from the University of Birmingham (Bryant Bequest; A.M.T and

866 Z.A.).

867

868 Competing interests

869 The authors report no competing interests.

870

35

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888 References

889 Ahmed Z, Aslam M, Lorber B, Suggate EL, Berry M, Logan A. Optic nerve and vitreal 890 inflammation are both RGC neuroprotective but only the latter is RGC axogenic. Neurobiol 891 Dis 2010a; 37(2): 441-54. 892 893 Ahmed Z, Bansal D, Tizzard K, Surey S, Esmaeili M, Gonzalez AM, et al. Decorin blocks 894 scarring and cystic cavitation in acute and induces scar dissolution in chronic spinal cord 895 wounds. Neurobiol Dis 2014; 64: 163-76. 896 897 Ahmed Z, Dent RG, Suggate EL, Barrett LB, Seabright RJ, Berry M, et al. Disinhibition of 898 neurotrophin-induced dorsal root ganglion cell neurite outgrowth on CNS myelin by siRNA- 899 mediated knockdown of NgR, p75NTR and Rho-A. Mol Cell Neurosci 2005; 28(3): 509-23. 900

36

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

901 Ahmed Z, Douglas MR, John G, Berry M, Logan A. AMIGO3 is an NgR1/p75 co-receptor 902 signalling axon growth inhibition in the acute phase of adult injury. 903 PLoS One 2013; 8(4): e61878. 904 905 Ahmed Z, Jacques SJ, Berry M, Logan A. Epidermal growth factor receptor inhibitors 906 promote CNS axon growth through off-target effects on glia. Neurobiol Dis 2009; 36(1): 142- 907 50. 908 909 Ahmed Z, Kalinski H, Berry M, Almasieh M, Ashush H, Slager N, et al. Ocular 910 neuroprotection by siRNA targeting caspase-2. Cell Death Dis 2011; 2: e173. 911 912 Ahmed Z, Read ML, Berry M, Logan A. Satellite glia not DRG neurons constitutively 913 activate EGFR but EGFR inactivation is not correlated with axon regeneration. Neurobiol Dis 914 2010b; 39(3): 292-300. 915 916 Ahmed Z, Read ML, Berry M, Logan A. Satellite glia not DRG neurons constitutively 917 activate EGFR but EGFR inactivation is not correlated with axon regeneration. Neurobiol Dis 918 2010c; 39(3): 292-300. 919 920 Ahmed Z, Suggate EL, Brown ER, Dent RG, Armstrong SJ, Barrett LB, et al. Schwann cell- 921 derived factor-induced modulation of the NgR/p75NTR/EGFR axis disinhibits axon growth 922 through CNS myelin in vivo and in vitro. Brain 2006; 129(Pt 6): 1517-33. 923 924 Almutiri S, Berry M, Logan A, Ahmed Z. Non-viral-mediated suppression of AMIGO3 925 promotes disinhibited NT3-mediated regeneration of spinal cord dorsal column axons. Sci 926 Rep 2018; 8(1): 10707. 927 928 Cai Y, Saiyin H, Lin Q, Zhang P, Tang L, Pan X, et al. Identification of a new RTN3 929 transcript, RTN3-A1, and its distribution in adult mouse brain. Brain Res Mol Brain Res 930 2005; 138(2): 236-43. 931 932 Chiurchiu V, Maccarrone M, Orlacchio A. The role of reticulons in neurodegenerative 933 diseases. Neuromolecular Med 2014; 16(1): 3-15. 934 935 Di Scala F, Dupuis L, Gaiddon C, De Tapia M, Jokic N, Gonzalez de Aguilar JL, et al. Tissue 936 specificity and regulation of the N-terminal diversity of reticulon 3. Biochem J 2005; 385(Pt 937 1): 125-34. 938 939 Fagoe ND, Attwell CL, Eggers R, Tuinenbreijer L, Kouwenhoven D, Verhaagen J, et al. 940 Evaluation of Five Tests for Sensitivity to Functional Deficits following Cervical or Thoracic 941 Dorsal Column Transection in the Rat. PLoS One 2016; 11(3): e0150141. 942 943 Farrukh F, Davies E, Berry M, Logan A, Ahmed Z. BMP4/Smad1 Signalling Promotes Spinal 944 Dorsal Column Axon Regeneration and Functional Recovery After Injury. Mol Neurobiol 945 2019. 946 947 Fawcett JW, Verhaagen J. Intrinsic Determinants of Axon Regeneration. Dev Neurobiol 948 2018; 78(10): 890-7. 949

37

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

950 Ferreira A, Niclas J, Vale RD, Banker G, Kosik KS. Suppression of kinesin expression in 951 cultured hippocampal neurons using antisense oligonucleotides. J Cell Biol 1992; 117(3): 952 595-606. 953 954 Fischer D, Hauk TG, Muller A, Thanos S. Crystallins of the beta/gamma-superfamily mimic 955 the effects of lens injury and promote axon regeneration. Mol Cell Neurosci 2008; 37(3): 471- 956 9. 957 958 Fournier AE, GrandPre T, Strittmatter SM. Identification of a receptor mediating Nogo-66 959 inhibition of axonal regeneration. Nature 2001; 409(6818): 341-6. 960 961 Futerman AH, Banker GA. The economics of neurite outgrowth--the addition of new 962 membrane to growing axons. Trends Neurosci 1996; 19(4): 144-9. 963 964 GrandPre T, Li S, Strittmatter SM. Nogo-66 receptor antagonist peptide promotes axonal 965 regeneration. Nature 2002; 417(6888): 547-51. 966 967 GrandPre T, Nakamura F, Vartanian T, Strittmatter SM. Identification of the Nogo inhibitor 968 of axon regeneration as a Reticulon protein. Nature 2000; 403(6768): 439-44. 969 970 Hains BC, Saab CY, Lo AC, Waxman SG. Sodium channel blockade with phenytoin protects 971 spinal cord axons, enhances axonal conduction, and improves functional motor recovery after 972 contusion SCI. Exp Neurol 2004; 188(2): 365-77. 973 974 Hata K, Fujitani M, Yasuda Y, Doya H, Saito T, Yamagishi S, et al. RGMa inhibition 975 promotes axonal growth and recovery after spinal cord injury. J Cell Biol 2006; 173(1): 47- 976 58. 977 978 Hirokawa N, Noda Y. Intracellular transport and kinesin superfamily proteins, KIFs: 979 structure, function, and dynamics. Physiol Rev 2008; 88(3): 1089-118. 980 981 Hirokawa N, Noda Y, Tanaka Y, Niwa S. Kinesin superfamily motor proteins and 982 intracellular transport. Nat Rev Mol Cell Biol 2009; 10(10): 682-96. 983 984 Hu X, Shi Q, Zhou X, He W, Yi H, Yin X, et al. Transgenic mice overexpressing reticulon 3 985 develop neuritic abnormalities. EMBO J 2007; 26(11): 2755-67. 986 987 Jacques SJ, Ahmed Z, Forbes A, Douglas MR, Vigenswara V, Berry M, et al. AAV8(gfp) 988 preferentially targets large diameter dorsal root ganglion neurones after both intra-dorsal root 989 ganglion and intrathecal injection. Mol Cell Neurosci 2012; 49(4): 464-74. 990 991 Kaplan A, Ong Tone S, Fournier AE. Extrinsic and intrinsic regulation of axon regeneration 992 at a crossroads. Front Mol Neurosci 2015; 8: 27. 993 994 Kimura T, Watanabe H, Iwamatsu A, Kaibuchi K. Tubulin and CRMP-2 complex is 995 transported via Kinesin-1. J Neurochem 2005; 93(6): 1371-82. 996 997 Kitchen P, Salman MM, Halsey AM, Clarke-Bland C, MacDonald JA, Ishida H, et al. 998 Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. 999 Cell 2020; 181(4): 784-99 e19.

38

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1000 1001 Konishi Y, Setou M. Tubulin tyrosination navigates the kinesin-1 motor domain to axons. Nat 1002 Neurosci 2009; 12(5): 559-67. 1003 1004 Kumamaru E, Kuo CH, Fujimoto T, Kohama K, Zeng LH, Taira E, et al. Reticulon3 1005 expression in rat optic and olfactory systems. Neurosci Lett 2004; 356(1): 17-20. 1006 1007 Lagord C, Berry M, Logan A. Expression of TGFbeta2 but not TGFbeta1 correlates with the 1008 deposition of scar tissue in the lesioned spinal cord. Molecular and cellular neurosciences 1009 2002; 20(1): 69-92. 1010 1011 Li S, Liu BP, Budel S, Li M, Ji B, Walus L, et al. Blockade of Nogo-66, myelin-associated 1012 glycoprotein, and myelin glycoprotein by soluble Nogo-66 receptor 1013 promotes axonal sprouting and recovery after spinal injury. J Neurosci 2004; 24(46): 10511- 1014 20. 1015 1016 Lo AC, Saab CY, Black JA, Waxman SG. Phenytoin protects spinal cord axons and preserves 1017 axonal conduction and neurological function in a model of neuroinflammation in vivo. J 1018 Neurophysiol 2003; 90(5): 3566-71. 1019 1020 Mahar M, Cavalli V. Intrinsic mechanisms of neuronal axon regeneration. Nat Rev Neurosci 1021 2018; 19(6): 323-37. 1022 1023 Matsuzaki F, Shirane M, Matsumoto M, Nakayama KI. Protrudin serves as an adaptor 1024 molecule that connects KIF5 and its cargoes in vesicular transport during process formation. 1025 Mol Biol Cell 2011; 22(23): 4602-20. 1026 1027 Mead B, Cullather E, Nakaya N, Niu Y, Kole C, Ahmed Z, et al. Viral delivery of multiple 1028 miRNAs promotes retinal ganglion cell survival and functional preservation after optic nerve 1029 crush injury. Exp Eye Res 2020; 197: 108071. 1030 1031 Mei F, Christin Chong SY, Chan JR. Myelin-based inhibitors of oligodendrocyte myelination: 1032 clues from axonal growth and regeneration. Neurosci Bull 2013; 29(2): 177-88. 1033 1034 Morgan-Warren PJ, O'Neill J, de Cogan F, Spivak I, Ashush H, Kalinski H, et al. siRNA- 1035 Mediated Knockdown of the mTOR Inhibitor RTP801 Promotes Retinal Ganglion Cell 1036 Survival and Axon Elongation by Direct and Indirect Mechanisms. Invest Ophthalmol Vis Sci 1037 2016; 57(2): 429-43. 1038 1039 Neumann S, Woolf CJ. Regeneration of dorsal column fibers into and beyond the lesion site 1040 following adult spinal cord injury. Neuron 1999; 23(1): 83-91. 1041 1042 Park KK, Liu K, Hu Y, Smith PD, Wang C, Cai B, et al. Promoting axon regeneration in the 1043 adult CNS by modulation of the PTEN/mTOR pathway. Science 2008; 322(5903): 963-6. 1044 1045 Raiborg C, Wenzel EM, Pedersen NM, Olsvik H, Schink KO, Schultz SW, et al. Repeated 1046 ER-endosome contacts promote endosome translocation and neurite outgrowth. Nature 2015; 1047 520(7546): 234-8. 1048

39

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1049 Read ML, Mir S, Spice R, Seabright RJ, Suggate EL, Ahmed Z, et al. Profiling RNA 1050 interference (RNAi)-mediated toxicity in neural cultures for effective short interfering RNA 1051 design. J Gene Med 2009; 11(6): 523-34. 1052 1053 Shi Q, Prior M, Zhou X, Tang X, He W, Hu X, et al. Preventing formation of reticulon 3 1054 immunoreactive dystrophic neurites improves cognitive function in mice. J Neurosci 2013; 1055 33(7): 3059-66. 1056 1057 Shirane M, Nakayama KI. Protrudin induces neurite formation by directional membrane 1058 trafficking. Science 2006; 314(5800): 818-21. 1059 1060 Stevens AR, Ahmed U, Vigneswara V, Ahmed Z. Pigment Epithelium-Derived Factor 1061 Promotes Axon Regeneration and Functional Recovery After Spinal Cord Injury. Mol 1062 Neurobiol 2019. 1063 1064 Surey S, Berry M, Logan A, Bicknell R, Ahmed Z. Differential cavitation, angiogenesis and 1065 wound-healing responses in injured mouse and rat spinal cords. Neuroscience 2014; 275: 62- 1066 80. 1067 1068 Tang BL. Protein trafficking mechanisms associated with neurite outgrowth and polarized 1069 sorting in neurons. J Neurochem 2001; 79(5): 923-30. 1070 1071 Tuxworth RIT, M.J.; Anduaga, A-M.; Hussien-Ali, A.; Chatzimatthaiou, S.; Longland, J.; 1072 Thompson, A.M.; Almutiri, S.; Alifragis, P.; Kyriacou, C.P.; Kysela, B.; Ahmed, Z. 1073 Attenuating the DNA damage response to double-strand breaks restores function in models of 1074 CNS neurodegeneration. Brain Communications 2019; 1(1): fcz005. 1075 1076 Vigneswara V, Akpan N, Berry M, Logan A, Troy CM, Ahmed Z. Combined suppression of 1077 CASP2 and CASP6 protects retinal ganglion cells from apoptosis and promotes axon 1078 regeneration through CNTF-mediated JAK/STAT signalling. Brain 2014; 137(Pt 6): 1656-75. 1079 1080 Vigneswara V, Berry M, Logan A, Ahmed Z. Caspase-2 is upregulated after sciatic nerve 1081 transection and its inhibition protects dorsal root ganglion neurons from apoptosis after serum 1082 withdrawal. PLoS One 2013; 8(2): e57861. 1083 1084 Wang X, Baughman KW, Basso DM, Strittmatter SM. Delayed Nogo receptor therapy 1085 improves recovery from spinal cord contusion. Ann Neurol 2006; 60(5): 540-9. 1086 1087 Yang YS, Strittmatter SM. The reticulons: a family of proteins with diverse functions. 1088 Biol 2007; 8(12): 234. 1089 1090 Yin Y, Henzl MT, Lorber B, Nakazawa T, Thomas TT, Jiang F, et al. Oncomodulin is a 1091 macrophage-derived signal for axon regeneration in retinal ganglion cells. Nat Neurosci 2006; 1092 9(6): 843-52. 1093 1094

1095

1096

40

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1097

1098

1099

1100

1101

1102

1103

1104

1105

1106

1107

1108

1109

1110

1111

1112

1113

1114 Figure 1. RTN3 is upregulated in dorsal root ganglion neurons (DRGN) after

1115 preconditioning lesions. (A) Diagram to show timeline of treatments and tissue

1116 harvesting. (B) Microarray data to show changes in RTN3 mRNA in DC, SN and

1117 pSN+DC lesions (n = 8 DRG/group (4 rats/group)). (C) qRT-PCR to confirm

1118 upregulated RTN3 mRNA in SN and pSN+DC injury models (n = 16 DRG/group (8

1119 rats/group)). (D) Western blot to show levels of levels of RTN3 in control, DC, SN and

1120 pSN+DC injury models (n = 16 DRG/group (8 rats/group)). (E) Densitometry to

1121 confirm upregulation of RTN3 in SN and pSN+DC injury models (n = 16 DRG/group

41

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1122 (8 rats/group)). (F) Immunohistochemistry to show immunolocalization of RTN3

1123 (green) in DRGN (red). Data are means ± SEM. Scale bars in (F) = 50µm. **P =

1124 0.001; ***P = 0.0001, one-way ANOVA with Dunnett’s post hoc test.

1125

1126 Figure 2. Overexpression of RTN3 in naïve DRGN cultures promotes neurite

1127 outgrowth. (A) Timeline of experiment and analysis methods. (B) Western blot to

1128 show RTN3 plasmids overexpress RTN3 protein. (C) Densitometry to show that

1129 RTN3 was significantly upregulated in RTN3 transfected cultures. (D) Representative

1130 images to show DRGN neurite outgrowth after RTN3 overexpression. RTN3

1131 overexpression increases (E) the mean DRGN neurite length, (F) % DRGN with

1132 neurites but does no affect (G) DRGN survival. LF2000 = Lipofectamine 2000, AU =

1133 arbitrary units, *** P =0.0001, one-way ANOVA with Dunnett’s post hoc test, n = 9

1134 wells/treatment. Scale bars in (D) = 100µm.

1135

1136 Figure 3. RTN3 is required for disinhibited DRGN neurite outgrowth. (A)

1137 Experimental timeline for analysis of RTN3 knockdown in preconditioned DRGN. (B)

1138 qRT-PCR to show ~80% knockdown of RTN3 using siRTN3. (C) Western blot to

1139 show levels of RTN3 protein after shRNA-mediated knockdown in DRGN. β-actin is

1140 used as a loading control. (D) Densitometry to show that PEI-shRTN3 significantly

1141 knocks down RTN3 protein in DRGN cultures. (E) Representative images to show

1142 neurite outgrowth after LF2000, siEGFP and siRTN3 treatment. (F) Quantification to

1143 show the mean longest neurite length after shRNA-mediated knockdown of RTN3.

1144 (G) Quantification to show % DRGN with neurites after shRNA-mediated knockdown

1145 of RTN3. (H) Quantification to show that DRGN survival is unaffected after shRNA-

42

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1146 mediated knockdown of RTN3. Data are means ± SEM. Scale bars in (E) = 100µm.

1147 ***P = 0.0001, one-way ANOVA with Dunnett’s post hoc test. n = 8 wells/condition.

1148

1149 Figure 4. Overexpression of RTN3 promotes disinhibited DRGN neurite

1150 outgrowth. (A) Timeline for experimental analysis of RTN3 overexpression in

1151 preconditioned DRGN. (B) Fold-change in mRNA after RTN3 overexpression in

1152 preconditioned DRGN. (C) Western blot to show overexpressed levels of RTN3

1153 protein. β-actin is used as a protein loading control. (D) Densitometry to show

1154 significant RTN3 overexpression by PEI-RTN3. (E) Representative images to show

1155 neurite outgrowth after DC+PEI-EGFP, pSN+DC+PEI-EGFP and pSN+DC+PEI-

1156 RTN3 treatment. (F) Quantification to show the mean neurite length after DC+PEI-

1157 EGFP, pSN+DC+PEI-EGFP and pSN+DC+PEI-RTN3 treatment (G) Quantification to

1158 show % DRGN with neurites after DC+PEI-EGFP, pSN+DC+PEI-EGFP and

1159 pSN+DC+PEI-RTN3 treatment. Data are means ± SEM. ***P = 0.0001, one-way

1160 ANOVA with Dunnett’s post hoc test. Scale bars in (E) = 100µm. n = 9

1161 wells/treatment.

1162

1163 Figure 5. Knockdown of Protrudin and its interactors attenuates RTN3-induced

1164 DRGN neurite outgrowth. (A) Timeline for experimental analysis of Protrudin and its

1165 interactors in RTN3-overexpressed DRGN. Representative western blots to confirm

1166 knockdown of (B) Protrudin, (C) FYCO1, (D) RAB7 and (E) SYT7. β-actin is used as

1167 a protein loading control. Representative images to show neurite outgrowth in (F)

1168 RTN3 overexpressed DRGN controls and after knockdown of (G) Protrudin, (H)

1169 FYCO1, (I) RAB7 and (J) SYT7 in RTN3-overexpressed cultures. (K) Quantification

1170 to show attenuation of RTN3-overexpressed DRGN neurite outgrowth by

43

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1171 shProtrudin, shFYCO1, shRAB7 and shSYT7. (L) DRGN survival remains unaffected

1172 by knockdown of Protrudin, FYCO1, RAB7 and SYT7 in RTN3-overexpressed

1173 DRGN. n = 9 wells/condition Scale bars in (F-J) = 100µm. Data are means ± SEM.

1174 ***P = 0.0001, one-way ANOVA with Dunnett’s post hoc test.

1175

1176 Figure 6. RTN3 overexpression promotes Protrudin-dependent DC axon

1177 regeneration. (A) Representative longitudinal sections of the spinal cord

1178 immunostained with GAP43 antibodies after treatment with pSN+DC+PEI-EGFP,

1179 pSN+DC+PEI-RTN3+shEGFP and pSN+DC+PEI-RTN3+shProtrudin (* = lesion site)

1180 (C to R = caudal to rostral). FluoroRuby (FR) traced axons in the same sections as

1181 pSN+DC+PEI-RTN3+shEGFP confirms overlap of GAP43+ axons (arrowheads),

1182 unequivocally demonstrating axon regeneration. Inset (i) shows high power images

1183 of GAP43+ axons in the boxed region (i). Inset (ii) and (iii) show high power images of

1184 FR+ axons in boxed regions (ii) and (iii). Note the overlap (arrowheads) between

1185 GAP43+ axons in inset (i) and FR+ axons in inset (ii). (B) Quantification of the % of

1186 GAP43+ axons at different distances caudal and rostral to the lesion site. Scale bars

1187 in (A) = 200µm. n = 18 rats/group. *P = 0.05, **P = 0.001, one-way ANOVA with

1188 Dunnett’s post hoc test.

1189

1190 Figure 7. RTN3 overexpression promotes electrophysiological, sensory and

1191 locomotor functional recovery after DC injury. (A) Representative Spike 2

1192 processed CAP traces in sham, DC+PEI-EGFP, pSN+DC+PEI-EGFP,

1193 pSN+DC+PEI-RTN3, pSN+DC+PEI-RTN3+shEGFP and pSN+DC+PEI-

1194 RTN3+shProtrudin-treated animals. (B) Negative CAP amplitudes at different

1195 stimulation intensities in sham, DC+PEI-EGFP, pSN+DC+PEI-EGFP, pSN+DC+PEI-

44

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1196 RTN3, pSN+DC+PEI-RTN3+shEGFP and pSN+DC+PEI-RTN3+shProtrudin-treated

1197 animals. (E) CAP areas at all stimulation intensities in sham, DC+PEI-EGFP,

1198 pSN+DC+PEI-EGFP, pSN+DC+PEI-RTN3, pSN+DC+PEI-RTN3+shEGFP and

1199 pSN+DC+PEI-RTN3+shProtrudin-treated animals. (F) Mean tape sensing and

1200 removal times in sham, DC+PEI-EGFP, pSN+DC+PEI-EGFP, pSN+DC+PEI-RTN3,

1201 pSN+DC+PEI-RTN3+shEGFP and pSN+DC+PEI-RTN3+shProtrudin-treated

1202 animals. (G) Mean error ratio to show the number of slips over the total number of

1203 steps in sham, DC+PEI-EGFP, pSN+DC+PEI-EGFP, pSN+DC+PEI-RTN3,

1204 pSN+DC+PEI-RTN3+shEGFP and pSN+DC+PEI-RTN3+shProtrudin-treated

1205 animals. Data are means ± SEM. n = 18 rats/group. ***P = 0.0001, one-way ANOVA

1206 with Dunnett’s post hoc test. #P = 0.0012, linear mixed models (LMM); ##P = 0.00012,

1207 generalised linear mixed models (GLMM).

1208

1209 Figure 8. RTN3 overexpression promotes RGC axon regeneration and

1210 functional recovery after ONC, which is also protrudin-dependent. (A) Western

1211 blot and (B) quantification to show that RTN3 plasmids significantly upregulate RTN3

1212 protein in the retina after intravitreal injection in vivo. (C) Immunohistochemistry to

1213 localise RTN3 protein to the RGCs (green) in the ganglion cell layer (GCL) and to the

1214 inner plexiform and inner nuclear layer (INL). ONL = outer nuclear layer. (D) High

1215 power images to show RTN3 protein localised to βIII-tubulin+ RGCs (arrows) in the

1216 GCL. (E) GAP43 immunohistochemistry and (F) quantification to show that in control

1217 optic nerve, few if any GAP43+ axon pass beyond the lesions site (*) whilst in RTN3

1218 overexpressed eyes, significant GAP43+ axons are present beyond the lesion site, an

1219 effect which is obliterated when Protrudin is knocked out at the same time as RTN3

1220 overexpression. (G) Representative images to show FluorGold (FG) backfilled RGC

45

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1221 in retinal wholemounts after RTN3 overexpression. (H) Quantification of the number

1222 of FG+ RGC in retinal wholemounts shows that overexpression of RTN3 is

1223 significantly neuroprotective, an effect which is independent of protrudin. (I)

1224 Representative ERG traces and (J) quantification of the pSTR amplitude shows

1225 significant improvements in RTN3 overexpressed eyes which is ablated after

1226 protrudin knockdown. Arrows show peak of pSTR. Data are means ± SEM. n = 12

1227 eyes/optic nerves/treatment. Scale bars in (C) = 100µm, scale bars in (D) = 25µm,

1228 scale bars in (E) = 200µm, scale bars in G = 50µm. ***P=0.0001, one-way ANOVA

1229 with Dunnett’s post hoc test.

1230

1231 Supplementary Figures

1232 Figure 3 – figure supplement 1. Knockdown of RTN3 mRNA by increasing

1233 amount of PEI-shRTN3 plasmid DNA in DRGN cultures. We determined that 2µg

1234 of shRTN3 plasmids were required for maximal RTN3 knockdown. Data are means ±

1235 SEM. n = 9 wells/treatment. ***P = 0.0001, one-way ANOVA with Dunnett’s post hoc

1236 test.

1237

1238 Figure 4 – figure supplement 1. Overexpression of RTN3 and challenge with

1239 Aβ1-42 does not have any adverse effects on DRGN neurite outgrowth or

1240 survival. (A) Immunostaining for RTN3 and βIII-tubulin in DRGN cultures, challenged

1241 with Aβ1-42 to show both upregulation of RTN3 in DRGN cultures and normal neurite

1242 outgrowth. High power images to show normal appearance of neurites in (B) control

1243 and (C) RTN3 overexpressed DRGN. (D) The mean neurite length, (E) % DRGN with

1244 neurites and (F) DRGN survival are unaffected by Aβ1-42 exposure. Scale bars in (A)

46

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1245 = 50µm, scale bars in (B and C) = 200 µm. ***P = 0.0001, one-way ANOVA with

1246 Dunnett’s post hoc test.

1247

1248 Figure 5 – figure supplement 1. Off-target gene expression is absent in RTN3-

1249 overexpressed DRGN treated with siRNAs. qRT-PCR shows lack of activation of

1250 interferon-related or innate immunity-related genes after siRNA transfections in

1251 RTN3-overexpressed DRGN. The positive control sequence (sip75NTR; Read et al.,

1252 2009) invokes 12-17-fold changes in all of these genes. Data are means ± SEM. n =

1253 6 wells/treatment.

1254

1255 Figure 6 – figure supplement 1. Overexpression of RTN3 and knockdown of

1256 Protrudin in DRGN in vivo. (A) Representative immunohistochemistry to show

1257 RTN3 immunoreactivity in control (pSN+DC+PEI-EGFP) and after overexpression of

1258 RTN3 in pSN+DC+PEI-RTN3-treated animals. (B) Western blot to show levels of

1259 RTN3 protein after overexpression of RTN3. β-actin is used as a protein loading

1260 control. (C) Densitometry showed significant upregulation of RTN3 protein after

1261 overexpression using RTN3 plasmids. (D) qRT-PCR to show knockdown in protrudin

1262 mRNA over time after in vivo DRG injection PEI-shProtrudin. Data are means ± SEM.

1263 n = 18 rats/group/test. Scale bars in (A) = 50µm. ***P = 0.0001, one-way ANOVA

1264 with Dunnett’s post hoc test.

1265

1266 Figure 7 – figure supplement 1. Overexpression of RTN3 in DRGN in vivo

1267 upregulates regeneration associated gene (RAGs) expression. (A)

1268 Immunohistochemistry to show induction of GAP43+ immunoreactivity in RTN3

1269 overexpressed DRGN (as measured by the reporter gene EGFP in green; arrows).

47

bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

1270 (B) qRT-PCR to demonstrate upregulation of RAGs (gap43, sprr1a, cebp-epsilon,

1271 atf3 and galanin) after preconditioning lesions and further upregulation after RTN3

1272 overexpression. Data are means ± SEM. n = 12 rats/group/test. Scale bars in (A) =

1273 25µm. ***P = 0.0001, one-way ANOVA with Dunnett’s post hoc test.

1274

1275 Figure 8 – figure supplement 1. RTN3 overexpression in retinal cultures

1276 promotes RGC survival and neurite outgrowth in vitro, which is dependent on

1277 protrudin. (A) Western blot to show that RTN3 is significantly overexpressed in

1278 retinal cells treated with RTN3 plasmids. (B) Densitometry shows greater than 3-fold

1279 upregulation of RTN3 protein levels in retinal cultures by RTN3 plasmids. (C)

1280 Representative images stained with βIII-tubulin and (D) quantification of the mean

1281 neurite length and (E) % RGC with neurites to show that RTN3 overexpression

1282 increased neurite length and the number of RGC with neurites, an effect which was

1283 ablated after knockdown of Protrudin. (F) RTN3 overexpression promotes significant

1284 RGC survival, which was not affected by protrudin knockdown. Data are means ±

1285 SEM. n = 9 wells/treatment. Scale bars in (C) = 100µm; ***P=0.0001, one-way

1286 ANOVA with Dunnett’s post hoc test.

1287

1288

48 bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. bioRxiv preprint doi: https://doi.org/10.1101/2021.05.24.445420; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.