Skip to main content
PLOS Biology logoLink to PLOS Biology
. 2022 Sep 20;20(9):e3001310. doi: 10.1371/journal.pbio.3001310

CBP/p300 activation promotes axon growth, sprouting, and synaptic plasticity in chronic experimental spinal cord injury with severe disability

Franziska Müller 1,#, Francesco De Virgiliis 1,#, Guiping Kong 1,#, Luming Zhou 1,#, Elisabeth Serger 1, Jessica Chadwick 1, Alexandros Sanchez-Vassopoulos 1, Akash Kumar Singh 2, Muthusamy Eswaramoorthy 3, Tapas K Kundu 2,4, Simone Di Giovanni 1,*
Editor: Cody J Smith5
PMCID: PMC9488786  PMID: 36126035

Abstract

The interruption of spinal circuitry following spinal cord injury (SCI) disrupts neural activity and is followed by a failure to mount an effective regenerative response resulting in permanent neurological disability. Functional recovery requires the enhancement of axonal and synaptic plasticity of spared as well as injured fibres, which need to sprout and/or regenerate to form new connections. Here, we have investigated whether the epigenetic stimulation of the regenerative gene expression program can overcome the current inability to promote neurological recovery in chronic SCI with severe disability. We delivered the CBP/p300 activator CSP-TTK21 or vehicle CSP weekly between week 12 and 22 following a transection model of SCI in mice housed in an enriched environment. Data analysis showed that CSP-TTK21 enhanced classical regenerative signalling in dorsal root ganglia sensory but not cortical motor neurons, stimulated motor and sensory axon growth, sprouting, and synaptic plasticity, but failed to promote neurological sensorimotor recovery. This work provides direct evidence that clinically suitable pharmacological CBP/p300 activation can promote the expression of regeneration-associated genes and axonal growth in a chronic SCI with severe neurological disability.


Spinal cord injury disrupts neural activity and is followed by a failure to mount an effective regenerative response. This study shows that pharmacological activation of CBP/p300 promotes histone acetylation and regenerative gene expression, counteracting retraction and promoting growth of sensory and motor axons in a mouse model of chronic spinal cord injury with severe disability.

Introduction

Spinal cord injury (SCI) is a devastating disease affecting millions of people worldwide. Severe SCI leads to permanent motor, sensory, and autonomic dysfunction that disrupts the quality of life of affected people. The management of severe SCI is nowadays limited to supportive care. Current physical rehabilitation has measurable but limited benefits after moderate SCI but fails to improve recovery after more severe and chronic injuries. This permanent loss of function is primarily caused by disruption of the connectivity of long-distance and intraspinal axonal fibres that fail to regenerate and reconnect with the neural circuitry below the injury [1]. To date, the lack of axonal regeneration after injury has been attributed to 2 main interconnected factors: (i) the formation of a cellular inhibitory environment that promotes growth cone collapse and (ii) the lack of an intrinsic regenerative response [2]. Furthermore, abnormal activity in spinal circuits below the injury contributes to a progressive deterioration of sensorimotor function [3] that is likely intensified in patients with long-standing chronic SCI. Therefore, neuromodulation/rehabilitation and axonal regrowth strategies seek to promote activity-dependent neuroplasticity to improve sensorimotor recovery.

Accumulating evidence suggest that increasing neuronal activity not only contributes to axonal sprouting, but it also strengthens synaptic plasticity and stimulates targeted axonal regrowth, favouring reconnectivity and functional recovery [4,5]. Modulation of axonal plasticity and growth of both motor and sensory fiber tracts, including of sensory circuits below the spinal lesion, can be carried out by specific neurorehabilitation schemes to enhance recovery after SCI [5]. However, this increase in regenerative growth and sprouting only partially promotes functional recovery, and it remains insufficient for reconnectivity and reestablishment of function in severe spinal injuries.

Attempts to stimulate the axonal regenerative response within the injured central nervous system (CNS) have been only partially successful through the manipulation of independent transcription factors or cofactors, including c-JUN, pCREB, SMAD1, MYC, HDAC5, KLF4, KLF7, and STAT3 [615]. Accumulating evidence shows that epigenetic modifications can contribute to the transcription-dependent enhancement of the regeneration programme in sensory axons or the injured optic nerve [1619]. Specifically, we found that the histone acetyltransferase (HAT) p300/CBP-associated factor (PCAF) acetylates the promoters of several regeneration-associated genes (RAGs) driving their expression after sciatic nerve injury and that PCAF overexpression promotes sensory axon regeneration across the injured spinal cord [18]. We and others have also shown that the HAT p300 can enhance optic nerve regeneration, promoting the expression of selected RAGs [17], while inhibiting class I histone deacetylases or HDAC3 partially promotes axonal regeneration of sensory axons following SCI [20]. However, modulation of these targets that were identified from injury-dependent paradigms have not translated into significant neurological recovery. We recently found changes in neuronal activity by housing mice in an enriched environment (EE) (large cages with toys, tunnels, running wheels, and enriched bedding) or following specific chemogenetic modulation of neural activity, induced epigenetic modifications, enabling active transcription, and regenerative growth. We next established that the CREB-binding protein (CBP) is the lysine acetyltransferase involved in this activity-dependent plasticity. Importantly, we showed that delivery of a small molecule specific activator of CBP/p300 named CSP-TTK21 promotes regenerative gene expression, axonal regeneration, plasticity, and functional sensorimotor improvement following acute SCI in rodents [21]. TTK21 is a HAT activator conjugated to a glucose-derived carbon nanosphere (CSP) able to cross the blood brain barrier, cell, and nuclear membranes with peak nuclear expression 3 days post-IP injection [22]. It was previously shown to promote neurogenesis and ameliorate memory deficits in tauopathy model in mice through increased histone acetylation and expression of genes involved in synaptic plasticity [22,23].

In addition, we have recently found that housing mice in an EE following a transection of the thoracic cord promotes significant sensorimotor recovery. These experiments suggest that housing animals in an EE might represent an “enriched” form of neurorehabilitation (S1 Fig). An alternative interpretation is that EE represents a more physiological environmental setting as opposed to standard housing (SH), which reflects an impoverished environment, especially when these housing conditions are compared to patients, suggesting that EE should be used as “standard” housing condition.

The lack of integration between approaches aimed to promote regenerative molecular mechanisms with neurorehabilitation and neuromodulation after SCI remains a major limitation for repair in severe and chronic SCI, where reawakening a regenerative gene expression programme and stimulation of disrupted neural activity are especially challenging and potentially critical to repair and recovery. Additionally, mechanistic and therapeutic advances in chronic SCI with severe disability are especially rare and therefore represent a high priority.

Hypothesis and relevance

Here, we hypothesize that the pharmacological stimulation of CBP/p300 activity will enhance regenerative gene expression during a growth refractory phase 12 weeks after spinal injury, while housing animals in an EE 1 week postinjury will stimulate neuronal activity, consolidate axonal and synaptic plasticity as opposed to animals housed in SH. Combined proteomics and transcriptomics studies indicate that EE activates physiological responses that are independent from CBP pathways as recently reported [21]. They include modulation of mitochondrial metabolism, calcium signalling, ion channels, axonal transport, and release of extracellular signalling vesicles among others, potentially allowing for synergistic benefits between the drug and EE.

Therefore, we postulate that CBP/p300 activator CSP-TTK21 on an EE housing baseline following the most clinically relevant chronic spinal cord injuries with severe disability might enhance neuronal plasticity, regeneration, and functional recovery, providing a better understanding of regenerative failure and filling a gap in the path to translation. Importantly, EE does not represent a specific form of focused rehabilitation, but rather a more physiological setting compared to SH that better reflects the human condition where patients are encouraged to engage in physical activities after a SCI. This explains the rationale for having animals in EE as baseline as opposed to having them in SH, which represents an impoverished artificial environment. Indeed, our study will test the effect of the CBP activator on this more “physiological” background in chronic SCI with severe disability.

The long-ranging implications of this work lie on testing a novel strategy based upon the CBP/p300 pharmacological activation to promote functional recovery after chronic SCI with severe disability, delivering the necessary preclinical evidence to support future clinical translation. Importantly, lack of validation of this hypothesis will also provide essential information allowing the scientific community to entertain alternative hypotheses. They include the possibility (i) that increases in regenerative gene expression and neural activity might not be synergistic in providing growth and plasticity; (ii) that recovery and repair might need task-specific neurorehabilitation; (iii) that reestablishment of functional circuitry needs targeting the spinal synapses; and finally (iv) that identification and manipulation of the extraneuronal wound healing and scarring processes in chronic SCI might need to be engaged along with favouring neuroplasticity.

Experimental design

The experimental design asked the question of whether activation of CBP and the cognate protein p300 with the weekly systemic delivery of TTK21 would enhance gene expression that supports plasticity and regenerative growth after chronic spinal cord transection injury with severe disability. Since no data were available on TTK21 in chronic SCI, a transection model in mice has been chosen versus a contusion in rats because (i) it allows a more accurate anatomical definition of axonal regeneration and sprouting; (ii) it has much less variability allowing for a more reliable statistical assessment keeping the number of animals relatively limited; and (iii) it needs a fraction of the drug required (in mice versus rats) that is produced in JNCASR, Bengaluru, India. Currently, the availability of the compound does not allow to initiate studies in rats in the short term; however, these studies will surely prompt further investigation in severe chronic contusion in rats, which is the next step towards human translation.

Specifically, adult 6- to 8-week-old C57Bl/6 mice received a spinal cord T9 transection injury that destroys the ascending sensory fibres in the dorsal columns bilaterally as well as most of the descending corticospinal, raphespinal, rubrospinal, and reticulospinal motor tracts, mimicking severe clinical SCI. This lesion leads to permanent severe impairment in sensorimotor function, severely limiting stepping up to at least 42 days postinjury, resembling clinical severe SCI (S1 Fig). All mice were placed in an EE 1 week after the SCI. The CBP activator TTK21 is coupled to slow-release carbon nanoparticles (CSP-TTK21) that allows a weekly administration maintaining stable activity levels in target tissues. Thus, CSP-TTK21 was administered via IP injections once per week starting 12 weeks after injury for 10 weeks. A control group of mice was treated with nanoparticles and vehicle alone (CSP). The CSP-TTK21 and control CSP nanoparticles were used at the dosage of 20 mg/kg that showed efficacy in subacute SCI as recently published [21].

Animals were killed at week 22 postinjury. Sprouting and regeneration of the dorsal columns were analyzed with the retrograde axonal tracer Dextran-488 (injected in the sciatic nerve in proximity of L4-L6 dorsal root ganglia (DRGs) 5 to 7 days before killing the animals). This allows visualization of ascending fibres in the dorsal columns into and across the injury site as previously shown in Di Giovanni’s lab [21]. Sprouting and regeneration of the corticospinal tracts (CSTs) were analysed with stereotaxic injections of the neural tracer Dextran-tetramethylrhodamine and biotin (Dextran T&B), into the motor cortex 2 weeks before killing, which allows visualization of descending fibres into and across the injury site according to standard procedures in Di Giovanni’s lab [24]. Axonal dieback as well as the number of fibres past the lesion site were normalized to the number of labelled fibres prior to the lesion as previously shown [24]. Given it closely correlates to locomotion, we also measured sprouting of serotoninergic raphe-spinal motor tracts with 5-HT immunohistochemistry as recently described [21]. To assess whether the CSP-TTK21 treatment enhances synaptic plasticity, we measured the number of inhibitory vGat or excitatory vGlut1 synaptic terminals in proximity of neuronal targets such as interneurons in the dorsal horns and motoneurons in the ventral horns of the spinal cord (ChAT or NeuN immunostaining) including in association with specific tracing of CST, sensory, or 5-HT fibres. This was carried out by fluorescent multilabelling experiments that were analyzed by confocal microscopy. Histone acetylation as read out of CBP/p300 activation was also evaluated in layer V neurons, raphe nuclei, and DRG neurons by immunofluorescence. The expression of several regeneration associated factors including ATF3, JUN, GAP43, SPRR1a, KLF7, pERK, and pSTAT3 was also studied by immunofluorescence in sensory and motor neurons.

In addition, we assessed locomotion, coordination, and sensorimotor integration by performing open field assessment with the Basso Mouse Scale (BMS) and the gridwalk tests, as recently shown [21]. Lastly, Von Frey test for mechanoception and mechanical allodynia as well as Hargreaves test for thermoception and thermal hyperalgesia were used to specifically assess the function of the ascending sensory tracts.

Please find a graphical summary (S2 Fig) and a summary table (Table 1) of the experimental design.

Table 1. Table summarizing the experimental design and data analysis.

Research question Hypothesis Sampling plan Statistical analysis Outcomes that confirm or disconfirm hypothesis
Does weekly systematic CSP-TTK21 delivery in a chronic severe SCI in mice housed in EE promote sensorimotor fiber regeneration? We hypothesize that treatment of CSP-TTK21 in mice housed in EE will enhance sensorimotor fiber regeneration due to the synergy between the TTK21 and EE enabling fibres to regenerate through the site of injury and provide directional regrowth.
Since EE is believed to enhance activity in both descending motor axons and facilitate motor control through the recruitment of proprioceptive feedback circuits, which are the pathways targeted by CBP/p300 activator, we expect a strong synergy between the drug and EE.
TTK21 vs. control: increase in % of axons regenerating beyond the lesion site from 5 to 10; SD = 2 and 4; Power 90%; P: 0.05, N: 8 For 5-HT immunostaining and for quantification of axonal regeneration of CST and dorsal column fibres:
Normally distributed data will be evaluated using a two-way repeated measures ANOVA with a Greenhouse-Geisser correction and a Tukey or Sidak post hoc test will be applied to examine multiple comparisons using a 95% confidence interval. For nonparametric evaluation, a Brunner & Langer nonparametric longitudinal data model could be used.
For VGlut and VGat quantification, an unpaired two-tailed Student t test with Welch correction and a 95% confidence interval will be applied. For nonparametric evaluation, a Mann–Whitney test will be used.
A threshold level of significance α was set at P value <0.05. Significance levels will be defined as follows: * P value <0.05; ** P value <0.01; *** P value <0.001, **** P value <0.0001. All data analysis will be performed blind to the experimental group.
Hypothesis would be confirmed if significant regeneration of sensory and motor (CST, raphespinal axons) and 5HT fibres are observed following CSP-TTK21 treatment compared to control.
CONFIRMED
Additional evidence to support this hypothesis would be provided by showing enhanced synaptic plasticity via a significant increase in the number of inhibitory VGat or excitatory VGlut synaptic terminals in proximity of neuronal targets in the spinal cord.
CONFIRMED for VGlut
NOT CONFIRMED for VGat
Does CSP-TTK21 systemic weekly delivery in mice housed in EE following a chronic and severe SCI enhance functional recovery? We hypothesize that treatment of CSP-TTK21 in mice housed in EE will promote functional recovery. Gridwalk: Control vs. TTK21: changes in score from 4 to 2; SD: 2 and 1; Power 90%; P: 0.05; N = 12 Gridwalk and BMS: Normally distributed data will be evaluated using a two-way repeated measures ANOVA and a Tukey or Sidak post hoc test will be applied to examine multiple comparisons using a 95% confidence interval. For nonparametric evaluation, a Brunner & Langer nonparametric longitudinal data model could be used.
A threshold level of significance α was set at P value <0.05. Significance levels will be defined as follows: * P value <0.05; ** P value <0.01; *** P value <0.001, **** P value <0.0001. All data analysis will be performed blind to the experimental group.
Hypothesis would be confirmed if significant differences were observed in the BMS and gridwalk scores between CSP-TTK21 and control. We would expect scores to remain similar until CSP-TTK21 injection, where we expect the CSP-TTK21 group to show an improvement in scores while no improvement is seen in the SP-Veh control.
NOT CONFIRMED
Does CSP-TTK21 systemic weekly delivery in mice housed in EE following a chronic and severe SCI promote gene expression and histone acetylation to enhance plasticity and regenerative growth? We hypothesize that stimulating CBP/p300 activity will enhance regenerative gene expression and histone acetylation during a refractory phase 12 weeks postinjury, while placing mice into EE 1 week postinjury stimulates neuronal activity and consolidates axonal and synaptic plasticity. TTK21 vs. control: change in fluorescence intensity of 1.5-fold; (between 50 and 75 arbitrary units, for example); SD = 10 and 12; P: 0.05; Power 90%; N = 4 IHC/IF: Normally distributed data will be evaluated using a two-tailed unpaired Student t test with Welch correction or a one-way ANOVA with a 95% confidence interval when experiments contained more than 2 groups. The Tukey post hoc test will be applied when appropriate. The Mann–Whitney U test will be used for nonparametric evaluation.
A threshold level of significance α was set at P value <0.05. Significance levels will be defined as follows: * P value <0.05; ** P value <0.01; *** P value <0.001, **** P value <0.0001. All data analysis will be performed blind to the experimental group.
Hypothesis would be confirmed if a significant increase in gene expression of regeneration associated genes (e.g. c-Jun, Atf3, pStat3) as well as increased histone acetylation (e.g. H3k27ac, H3k9ac, H4k8ac) was observed following CSP-TTK21 treatment versus control.
CONFIRMED for selected genes

BMS, Basso Mouse Scale; CST, corticospinal tract; EE, enriched environment; IF, immunofluorescence; IHC, immunohistochemistry; SCI, spinal cord injury.

Results

CBP/p300 activator TTK21 promotes histone acetylation and the expression of regeneration-associated signals in neurons in chronic SCI

Delivery of the CBP/p300 activator TTK21 promoted histone acetylation as expected in DRG (Fig 1A–1D), raphe (Fig 1E and 1F), and layer V cortical neurons (Fig 1G–1J) at 22 weeks after SCI. Further, as shown by immunofluorescence experiments, TTK21 enhanced the expression of ATF3, SPRR1a, cJUN, KLF7, and GAP43 (Fig 2A–2J), however not of pERK and pSTAT3 (Fig 2K–2N) in DRG neurons. ATF3, KLF7, pSTAT3, and pERK were not expressed above background in layer V cortical neurons, while cJUN and SPRR1a were unaffected by TTK21 (S3 Fig). Thus, CBP/p300 activation promotes histone acetylation in both motor and sensory neurons as well as the expression of RAGs in sensory DRG neurons in chronic SCI.

Fig 1. Histone acetylation in DRG, raphe, and layer 5 cortical neurons in CSP-TTK21-treated mice in a chronic SCI with severe disability.

Fig 1

(A) Representative micrographs of H3K9ac immunostaining (green, white arrows) in DRG neurons. (B) Quantification of H3K9ac immunostaining in DRG neurons from CSP or CSP-TTK21-treated mice (CSP: 3,505.0 ± 399.8; CSP-TTK21: 6,549.0 ± 122.1, p < 0.001, n = 4). (C) Representative micrographs of H3K27ac immunostaining (green, white arrows) in DRG neurons. (D) Quantification of H3K27ac immunostaining in DRG neurons in CSP or CSP-TTK21-treated mice (CSP: 7,738.0 ± 472.0, n = 5; CSP-TTK21: 9,779.0 ± 195.4, p < 0.01, n = 4). (E) Representative micrographs of H3K9ac staining (green, white arrows) in raphe neurons. (F) Quantification of H3K27ac immunostaining in raphe neurons from CSP or CSP-TTK21-treated mice (CSP: 15.9 ± 0.2; CSP-TTK21: 27.1 ± 1.4, p < 0.001, n = 4). (G) Representative micrographs of H3K9ac immunostaining (green, white arrows) in layer 5 cortical neurons. (H) Quantification of H3K9ac immunostaining in layer 5 cortical neurons from CSP or CSP-TTK21-treated mice (CSP: 18.2 ± 1.8; CSP-TTK21: 39.0 ± 4.1, p < 0.01, n = 4). (I) Representative micrographs of H3K27ac staining (green, white arrows) in layer 5 cortical neurons. (J) Quantification of H3K27ac immunostaining in layer 5 cortical neurons from CSP or CSP-TTK21-treated mice (CSP: 22.9 ± 2.9; CSP-TTK21: 42.1 ± 1.3, p < 0.001, n = 4). Mean ± SEM; unpaired two-tailed Student t test; ** p < 0.01, *** p < 0.001. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; DRG, dorsal root ganglion; SCI, spinal cord injury.

Fig 2. Expression of regeneration-associated proteins in DRG neurons in CSP-TTK21-treated mice in a chronic SCI.

Fig 2

(A-N) Representative micrographs of immunostaining for regeneration-associated proteins (green, white arrows) in DRG neurons from CSP or CSP-TTK21-treated mice (A, C, E, G, I, K, M) and respective quantification of immunostaining in DRG neurons (B, D, F, H, J, L, N). (A, B) Atf3 (CSP: 2,113.0 ± 461.5; CSP-TK21: 4,614.0 ± 449.4, p < 0.05). (C, D) Sprr1a (CSP: 4,904.0 ± 1,033.0; CSP-TK21: 9,331.0 ± 914.6, p < 0.05). (E, F) cJun (CSP: 3,697.0 ± 302.2; CSP-TK21: 4,904.0 ± 340.7, p < 0.05). (G, H) Klf7 (CSP: 4,097.0 ± 199.1; CSP-TK21: 772.00 ± 317.9, p < 0.0001). (I, J) Gap43 (CSP: 1,178.0 ± 173.2; CSP-TK21: 4,643.0 ± 163.6, p < 0.001). (K, L) pErk (CSP: 3,433.0 ± 428.9; CSP-TK21: 3,036.0 ± 750.7, p = 0.66, TOST: t(4.8) = 0.03, p = 0.51 given equivalence bounds of −366.7 and 366.7 on a raw scale and alpha of 0.05). (M, N) pStat3 (CSP: 2,894.0 ± 470.1; CSP-TK21: 3,579.0 ± 656.8, p = 0.43, TOST: t(5.4) = −0.42, p = 0.66 given equivalence bounds of −342.7 and 342.7 on a raw scale and alpha of 0.05). Mean ± SEM; unpaired two-tailed Student or Welch t test; *p < 0.05, *** p < 0.001, **** p < 0.0001. n = 3 or 4 biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; DRG, dorsal root ganglion; SCI, spinal cord injury; TOST, two one-sided tests.

CBP/p300 activator TTK21 enables axonal growth and synaptic plasticity in chronic SCI

Delivery of the CBP/p300 activator TTK21 significantly reduced axonal retraction and promoted axonal growth of motor corticospinal (Fig 3A and 2B) and of sensory DRG neurons, respectively (Fig 3C and 3D). However, the most striking phenotype was the increased sprouting of 5-HT raphe-spinal axons as shown by 5-HT immunofluorescence (Fig 3E and 3F). Importantly, TTK21 led to an increased number of vGlut1, but not of vGat, boutons opposed to motor neurons in the ventral horn of L1-3 spinal sections below the injury site (Fig 3G–3L).

Fig 3. Axonal growth and synaptic plasticity in CSP-TTK21-treated mice in a chronic SCI.

Fig 3

(A) Representative micrographs of BDA-traced CST axons (green, white arrows) after chronic SCI in CSP or CSP-TTK21-treated mice, GFAP (magenta) was used to determine the lesion site. (B) Average distance between the lesion border and the furthest rostral (CSP: −329.8 ± 41.4, n = 6; CSP-TTK21: 438.3 ± 40.4, p < 0.0001, n = 9) or caudal axons (CSP: −548.6 ± 58.7, n = 7; CSP-TTK21: −366.2 ± 54.6, p < 0.05, n = 9) in CSP and CSP-TTK21-treated mice. (C) Representative micrographs of Dextran-traced sensory axons (green, white arrows) after chronic SCI in CSP or CSP-TTK21-treated mice. GFAP (magenta) was used to determine the lesion site. (D) Average distance between the lesion border and furthest rostral (CSP: −361.2 ± 47.3, n = 6; CSP-TTK21: 301.6 ± 35.8, p < 0.0001, n = 5) or caudal axons (CSP: −610.5 ± 25.2, n = 6; CSP-TTK21: −227.8 ± 22.7, p < 0.0001, n = 7) in CSP and CSP-TTK21-treated mice. (E) Representative micrographs of 5-HT-positive axons (green) sprouting around ChAT-positive motoneurons (magenta) in the lumbar ventral horn (white dotted lines) below the lesion site in CSP or CSP-TTK21-treated mice. (F) Quantification of 5-HT intensity in the lumbar ventral horn (CSP: 2,410.0 ± 549.3; CSP-TTK21: 5,189.0 ± 662.2, p < 0.05, n = 4). (G) Orthogonal 3D confocal images of VGlut1+ boutons (green) from group-1a afferents in proximity to motoneurons (ChAT+, magenta) in the lumbar ventral horn below the lesion site. Intersection of dotted lines indicates example of apposition. (H) Quantification of Vglut1+ puncta per 1 mm2 in CSP or CSP-TTK21 mice (CSP: 6,380.0 ± 483.1, n = 3, CSP-TTK21: 11,615.0 ± 1,337.0, n = 4, p < 0.05). (I) Quantification of VGlut1+ boutons in proximity to motoneurons (CSP: 1.7 ± 0.2; CSP-TTK21: 4.5 ± 0.3, p < 0.0001, n = 4). (J) Orthogonal 3D confocal images of VGat+ boutons (green) in proximity to motoneurons (ChAT+, magenta) in the lumbar ventral horn below the lesion site. Intersection of dotted lines indicates example of apposition. (K) Quantification of VGat+ puncta per 1 mm2 (CSP: 66,366.0 ± 2,970.0; CSP-TTK21: 66,996.0 ± 2,069.0, p = 0.87, TOST: t(5.4) = 0.2, p = 0.41 given equivalence bounds of −1,535.7 and 1,535.7 on a raw scale and alpha of 0.05, n = 4). (L) Quantification of VGlut1+ boutons in proximity to motoneurons (CSP: 7.0 ± 1.1; CSP-TTK21: 9.3 ± 0.9, p = 0.15, TOST: t(5.7) = −1.2, p = 0.86 given equivalence bounds of −0.6 and 0.6 on a raw scale and alpha of 0.05, n = 4). All data are given as mean ± SEM; unpaired two-tailed Student t test; * p < 0.05, *** p < 0.001, **** p < 0.0001. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; CST, corticospinal tract; SCI, spinal cord injury; TOST, two one-sided tests.

No difference in the size of the lesion and in GFAP astrocytic immunostaining was observed between the experimental groups (S4A–S4C Fig). Similarly, measurement of macrophage/microglia CD68 immunofluorescence intensity around the injury did not show any difference between vehicle and TTK21 (S5 Fig), suggesting that systemic activation of CBP/p300 does not lead to obvious changes in the spinal cord astrocytic and macrophage/microglia responses. Thus, CBP/p300 activation can promote axonal growth and synaptic plasticity in chronic SCI with severe disability.

CBP/p300 activator TTK21 does not affect neurological recovery in chronic SCI

Lastly, we measured whether CBP/p300 activation promotes sensorimotor recovery in chronic SCI. We did not observe any significant difference in the sensorimotor performance by BMS (Fig 4A) and gridwalk (Fig 4B) between TTK21 and vehicle-treated animals, while the severity of the lesion was confirmed by the severe impairment observed (Fig 4C). Only a minority of animals were able to perform stepping on a gridwalk (BMS >3; Fig 4B). Similarly, the animals that could be tested for mechanoception by Von Frey and thermal responses by Hargreaves showed severe impairments with no difference between the 2 experimental groups (Fig 4D and 4E). Thus, CBP/p300 activation cannot stimulate sensory or motor recovery with this level of chronic SCI.

Fig 4. Sensorimotor behavioural tests after CSP-TTK21 treatment in chronic SCI with severe disability.

Fig 4

(A) BMS quantification of CSP (n = 15) or CSP-TTK21 (n = 14) treated mice after chronic SCI (Treatment: f(1) = 0.002, p = 0.96 (TOST: given equivalence bounds of −0.21 and 0.21, 90% confidence intervals fall −0.37 and 0.32 and p = 0.307, thus H0 is undecided); Time: f(12) = 192.1, p < 0.001 (TOST: given equivalence bounds of −0.21 and 0.21, 90% confidence intervals fall −0.24 and −0.15 and p = 0.215, thus H0 is rejected); Interaction (treatment × time): f(12) = 6.6, p = 0.82 (TOST: given equivalence bounds of −0.21 and 0.21, 90% confidence intervals fall 4.19 and 4.99 and p > 0.999, thus H0 is rejected). Two-way repeated measures ANOVA with Sidak post hoc test. (B) Gridwalk quantification of the percentage of slips per total number of steps per run in CSP (n = 4) or CSP-TTK21 (n = 6) treated mice with a BMS score greater than 3 after chronic SCI (Treatment: f(1) = 0.2, p = 0.69 (TOST: given equivalence bounds of −1.78 and 1.78, 90% confidence intervals fall −7.96 and 2.63 and p = 0.693, thus H0 is undecided); Time: f(12) = 7.1, p < 0.001 (TOST: given equivalence bounds of −1.78 and 1.78, 90% confidence intervals fall −0.73 and 0.66 and p < 0.001, thus H0 is accepted); Interaction (treatment × time): f(12) = 6.6, p = 0.82 (TOST: given equivalence bounds of −1.78 and 1.78, 90% confidence intervals fall −28.59 and 41.29 and p > 0.999, thus H0 is rejected). Two-way repeated measures ANOVA with Sidak post hoc test). (C) Bar graph indicating the number of mice with a BMS score greater than 3 across postinjury time points. (D) Hargreaves test indicating average paw withdrawal latency in CSP or CSP-TTK21 (Welch two-tailed t test: CSP: 6.7 ± 1.1, n = 4; CSP-TTK21: 7.6 ± 0.5, n = 6, p = 0.51, TOST: t(8) = −0.3, p = 0.63 given equivalence bounds of −0.5 and 0.5 on a raw scale and an alpha of 0.05) treated mice with a BMS score greater than 3. (E) Von Frey test indicating average paw withdrawal threshold in CSP or CSP-TTK21 (Welch two-tailed t test: CSP: 5.5 ± 1.6, n = 4; CSP-TTK21: 6.4 ± 0.7, n = 6, p = 0.64, TOST: t(4.3) = −0.1, p = 0.52 given equivalence bounds of −0.8 and 0.8 on a raw scale and an alpha of 0.05) treated mice with a BMS score greater than 3. All data are given as mean ± SEM. n = biologically independent animals. The data can be found in S1 Data. BMS, Basso Mouse Scale; CSP, carbon nanosphere; SCI, spinal cord injury; TOST, two one-sided tests.

Discussion

Chronic severe SCI that is associated with absent axonal regrowth and reconnectivity and severe neurological disability remains a major medical challenge. It is believed that the ability for axonal growth and sprouting declines over time after an SCI and that the time window for intervention might eventually close. The importance of our findings lies on the evidence that starting a treatment such as the CBP/p300 activator TTK21 12 weeks after an SCI in the mouse with severe disability can still elicit a regenerative response as shown by increased axonal growth, sprouting, and synaptic plasticity at 22 weeks postinjury. The CBP/p300 activator TTK21 also promotes the expression of regenerative signals including some well-established RAGs. This is a potentially exciting discovery since it provides a demonstration that a clinically suitable molecular intervention can promote plasticity and growth in both an acute, as previously shown [21], and chronic SCI with severe disability, likely by reawakening a dormant regenerative gene expression programme. While both motor and sensory neurons showed increased histone acetylation, CBP/p300 activation induced the expression of most RAGs investigated in DRG neurons only. These findings are in line with previous work in subacute SCI where these classical RAGs were not activated in the corticospinal [25], which temporarily revert to an embryonic state, as opposed to DRG neurons, which reexpress RAGs, as shown here. However, the increase in histone acetylation seems to be a common denominator of TTK21 delivery for sensory and motor neurons alike. Increased acetylation likely stimulates distinct regenerative programmes between peripheral sensory and central motor neurons by enhancing chromatin accessibility at gene regulatory regions of RAGs for sensory neurons and perhaps of developmentally regulated genes for corticospinal neurons as recent work from the Tuszynski’s lab might suggest [25]. They found that reexpression of embryonic genes after SCI lasts for 2 weeks only in corticospinal neurons. In the present chronic state of injury (12 to 22 weeks after injury), this “primed” state for regeneration has likely closed. Thus, delivery of CSP-TTK21 might have partially reopened this regenerative window. It is in fact important that TTK21 increases the growth and regenerative gene expression ability of both sensory and motor neurons, albeit with differential potency and efficacy, being maximal for regenerative gene expression at selected RAGs in sensory DRG neurons and for axonal sprouting in 5-HT motor neurons. The very distinct embryonic origin and molecular identities of these CNS neuronal subpopulations might underline this differential response [26]. However, the variable distance from the lesion site and the rate of axonal transport of selected neuronal populations might be additional contributing factors since neuronal cell types and the distance of the neuronal cell body from the injury site affect the rate and extent of axonal trafficking and the expression of RAGs.

Given the severity of the spinal injury in the present study, it is very difficult to directly compare these findings with the level of axonal growth, sprouting, and synaptic plasticity observed in our previous work when we delivered TTK21 6 or 24 hours after a mouse spinal cord hemisection or a rat moderate to severe contusion injury, respectively [21]. However, the most striking difference between the present and the previous experiments is the lack of any neurological recovery. The most immediate explanation might be the modest effect of axonal growth of sensory and corticospinal neurons, the depth of the transection injury in the present study compared to a dorsal hemisection or a moderate to severe contusion in the previous. The presence of astrocytic rich tissue bridges is important to allow for axonal growth, and the present chronic injury showed poor tissue bridges. Additionally, the diverse injury severities and timing postinjury likely reflect a different glial environment that might influence synaptic transmission, repair, and functional recovery. Finally, in a chronic condition, synaptic transmission might be compromised by the functional impairment of neuronal targets below (motor targets) or above (sensory targets) the injury site due to the long-standing deafferentation.

However, our positive neuroanatomical and molecular findings despite the limitations of the lack of neurological recover might pave the way for the future combinatorial use of TTK21 activation with stem cell grafts [25,27], self-assembling peptide-based biomaterials [28], or both, since these might provide the necessary tissue bridge and relays to allow for increased functional synapses underpinning neurological recovery. Additionally, while in our view, tissue bridges will need to be part of the equation for spinal repair, the synergism with additional neuronal extrinsic interventions aimed to further enhance plasticity such as CSPG inhibition could also be considered.

More broadly, extending upon previous findings showing that PTEN deletion [29] or antagonism [30] benefit axonal growth and spinal circuitry formation in chronic SCI, the present work suggests that it is possible to promote axonal growth and plasticity in a chronic spinal cord injury leading to severe disability. Finally, similarly to TTK21, it encourages to attempt other molecular regenerative interventions, ideally in combination with tissue bridging approaches.

Materials and methods

Mice

Mouse experimentation was carried out in accordance with regulations of the UK Home Office under the Animals (Scientific Procedures) Act 1986, with Local Ethical Review by the Imperial College London Animal Welfare and Ethical Review Body Standing Committee (AWERB, PPL P6EDD65B1). Female C57Bl6 (Harlan, UK) mice ranging from 6 to 8 weeks of age were used for all experiments. For all surgeries, mice were anesthetized with isoflurane (4% induction 2% maintenance) and buprenorphine (0.1 mg/kg) and carprofen (5 mg/kg) were administered peri-operatively as analgesic.

Animals were kept on a 12-hour light/dark cycle with food and water provided ad libitum, at a constant room temperature and humidity (21°C and 5%, respectively). SH for mice consists of 26 × 12 × 18 cm3 cages housing 4 mice with tissue paper for bedding, a tunnel, and a wooden chew stick. The EE housing consists of 36 × 18 × 25 cm3 cages housing 5 mice with tissue paper for bedding, a tunnel, and a wooden chew stick. EE cages also host the following: additional nesting material that included nestlets, rodent roll, and sizzle pet (LBS Biotech); a hanging plastic tunnel (LBS Biotech) and a plastic igloo combined with a fast-track running wheel (LBS Biotech); a wooden object (cube, labyrinth, tunnel, corner 15) (LBS Biotech) that is changed every 5 days to help maintain a novel environment; and 15 g fruity gems (LBS Biotech) every 5 days to encourage exploratory and natural foraging behaviour.

Spinal cord injury (SCI)

Surgeries were performed as we previously reported [24]. A laminectomy at vertebra T9 was performed to expose spinal level T9 and a deep dorsal transection past the central canal at a depth of approximately 1.1 mm was carried out using microscissors (Fine Science Tools). This lesion only spares a portion of the ventral white matter and leaves the animals with a severe and permanent neurological impairment—no improvement from baseline. At week 22 after SCI, animals were deeply anesthetized and perfused transcardially.

CSP-TTK21 administration

Animals were randomised to treatment after been subdivided into 2 groups with comparable severity based upon BMS with the CBP/p300 activator bound to carbon nanospheres (CSP-TTK21) or a control of just CSP (mice– 20 mg/kg injected IP once a week). Mice received the first IP injection 12 weeks after SCI until killing.

Neuronal tracing

For dorsal column tracing of sensory DRG axons, 2 μl of Dextran-88 (Life Technology) was injected into the sciatic nerve bilaterally 1 week before killing using the fire polished glass capillary connected to a 10-μl Hamilton syringe.

For anterograde tracing of motor CSTs, 2 weeks before killing, mice that received an SCI were injected with the axonal tracer Dextran-tetramethylrhodamine and biotin, with a Hamilton microsyringe in the motor cortex following standard stereotaxic coordinates as previously described [31].

5-HT immunostaining

For 5-HT immunostaining, we followed a protocol we previously described [32]. The sections were incubated with rabbit anti-5-HT (1:500, Sigma S5545) in 4% NGS in 0.3% TBS-Triton X100 for 4 days at 4°C. Next, the sections were incubated with an Alexa fluorescent secondary antibody. Finally, the sections were coverslipped.

Behavioral analysis

Mice were trained daily for 2 weeks preinjury before baseline measurements and then assessed on day 7 postinjury and biweekly thereafter; until CSP or CSP-TTK21 injections, mice were then tested weekly thereafter. All behavioral testing and analysis were done by 2 observers blinded to the experimental groups.

Gridwalk

Mice will cross a 1-m long horizontal grid 3 times. Videos of the runs were blindly analyzed at a later time point, and errors from both hind limbs were counted and normalized to total number of steps. Error values represent the total number of slips made by both hindlimbs over the 3 runs.

Open-field test

The BMS [33] was used to assess open-field locomotion. Each animal was allowed to freely move in the open field for 4 minutes while 2 independent investigators blinded to experimental group will score it. The BMS score and subscore were given. Only the animals showing frequent or consistent plantar stepping in the open field (BMS score ≥3) were tested on the grid walk.

Von Frey

The Von Frey test determines the mechanical force required to elicit a paw withdrawal response. Each animal was tested in each paw 3 times. Only animals showing plantar placement in the open field (BMS score ≥3) were tested.

Hargreaves

The Hargreaves test determines the latency of a thermal nociceptive stimulus required to elicit a paw withdrawal response. Each animal was in each paw 3 times. Only animals showing plantar replacement in the open field (BMS score ≥3) were tested.

Quantification of axonal regeneration

For each spinal cord after injury, the number of fibres rostral and caudal to the lesion and their distance from the lesion epicentre (depending on whether sensory or motor axons) were analysed in 4 to 6 sections per animal with a fluorescence Axioplan 2 (Zeiss) microscope and with the software Stereo-Investigator 7 (MBF Bioscience). The lesion epicentre was identified by GFAP staining in each section at 20× magnification. The total number of labelled axons or signal intensity of the traced axons rostral to the lesion site were normalized to the total number of labelled axons or of the signal intensity caudal or rostral to the lesion site counted in all the analysed sections for each animal, obtaining an inter-animal comparable ratio. Sprouts and regrowing fibres were defined following the anatomical criteria reported by Steward and colleagues [34].

Histology and immunohistochemistry

Tissue was postfixed in 4% paraformaldehyde (PFA) (Sigma) and transferred to 30% sucrose (Sigma) for 5 days for cryoprotection, the tissue was then be embedded in OCT compound (Tissue-Tek) and frozen at −80°C. DRG, spinal cord, and cortices were sectioned at 10, 20, and 20 μm thickness, respectively, using a cryostat (Leica). Immunohistochemistry on tissue sections was performed according to standard procedures. For selected antibodies, antigen retrieval was performed submerging the tissue sections in 10 mM citrate buffer (pH 6.2) or 10 mM Tris/1 mM EDTA buffer (pH 9.0) at 98°C for 5 minutes. Next, tissue sections were washed with PBS to remove the excess of citrate buffer and blocked for 1 hour with 8% BSA, 1% PBS-TX100. Finally, the sections were incubated with anti-p-STAT3 (1:100, Rabbit, Cell Signaling Technology 9145), c-Jun (1:100, CST, #9165), ATF3 (1:100, Santa Cruz, sc-188), pErk (1:250, CST, #9101), GAP43 (1:500, Sigma AB5220), KLF7 (1:100, Santa Cruz sc-398576), SPRR1A (1:100, Thermo Fisher Scientific PA5-110423), Tuj1 (1:1,000, Promega G7121), H3K27ac (1:500, ab4729), H3K9ac (1:500, Cell Signalling 9671), GFAP (1:500, Millipore AB5804), CD68 (1:500, Abcam ab213363), vGlut1 (1:1,000, Synaptic system 135302), vGat1 (1:500, Synaptic systems 131011), ChAT (1:500, Sigma AB144P) antibodies at 4°C O/N. This was followed by incubation with Alexa Fluor–conjugated secondary antibodies according to standard protocol (1:1,000, Invitrogen). Slides were counterstained with DAPI to visualise nuclei whenever necessary (1:5,000, Molecular Probes).

Image analysis for IHC

All analysis was performed by the same experimenter who was blinded to the experimental groups. Photomicrographs were taken with a Nikon Eclipse TE2000 microscope with an optiMOS scMOS camera using 10× or 20× magnification using ImageJ (Fiji 64 bits 1.52 p), Micro-Manager 2.0 software for image acquisition or at 20× magnification with an Axioplan 2 (Zeiss) microscope and processed with the software AxioVision (Zeiss).

Analysis of GFAP and CD68 intensity around the lesion site

GFAP and CD68 intensity and area with positive signal were quantified from sagittal spinal cord sections from 1 series of tissue for each animal. Quantification was done using ImageJ, the background was subtracted, and then the mean pixel intensity and area of immunoreactivity was measured.

Analysis of fluorescence intensity

For quantitative analysis of pixel intensity, the nucleus or soma of DRG or layer V cortical neurons were manually outlined in images from 1 series of stained tissue for each mouse. To minimize variability between images, pixel intensity was normalized to an unstained area and the exposure time and microscope setting were fixed throughout the acquisition.

Analysis of 5HT fibres in the ventral horn

Intensity of 5HT immunohistochemistry was measured in the ventral horn of L1-4 spinal sections. Quantification was done using ImageJ, the background was subtracted, and then the mean pixel intensity was measured from 1 series of tissue for each animal.

Analysis of vGlut1 and vGat immunohistochemistry in proximity to motor neurons

vGlut1 and vGat synaptic boutons were imaged with a SP8 Leica confocal microscope. Z-stacks images were taken with an average thickness of 15 μm with a step size of 0.3 μm. Sequential line scanning was performed when more than 2 channels were acquired. Multifluorescent orthogonal 3D image analysis and visualization were performed using Leica LAS X software. The average number of vGlut1 or vGat boutons opposed to motor neurons in the ventral horn of L1-3 spinal sections was calculated by analysing 20 motor neurons per replicate. All analyses were performed blind to the experimental group.

Statistical analysis

Measures for avoidance of bias (e.g., blinding, randomisation)

We adhere to the principles of NC3Rs and adhere to the ARRIVE guidelines [35]. All experiments were performed in blind to the treatment and experimental group. Behavioral studies were assessed by 2 independent investigators (e.g., research associate and research assistant) blind to one another, to the treatment, and to injury group when relevant. Randomization followed a computerized sequence. As far statistical analysis, our priority was to adopt a frequentist equivalence test. There are 5 parameters in our experimental design: sample size (n), difference between the groups (delta), standard deviation (sigma), type I error (alpha/significance), and type II error (power). We can use estimates for 4 of these parameters to calculate the 5th parameter—which we have done to achieve our required sample size to achieve a certain power and significance.

Sample size calculation, power calculations, and justification of effect size

The size of our in vivo experimental groups as planned in our aims has been defined following the Animal Experimentation Sample Size Calculator (AESSC) tool. Sample size calculations were performed using a two-tailed unpaired Student t test and two-way repeated measures ANOVA (significance ≤ 0.05; power ≥ 0.90; G*Power); N indicates number/group. The specific effect size has been estimated based upon similar studies showing significant differences between experimental and control group [8,21].

Two animals were added to each experiment based upon the probability of losing animals due to the experimental procedure such as spinal surgery. While the selected N is derived by our power calculation, it is also compatible and comparable with what we have previously published for similar experiments [21]. Exclusion criteria include the following:

  • Tissues with low quality for further experimentation or imaging as shown by lack of clear injury site or by lack of clear axonal tracing. Degraded cords in case of inefficient fixation or damaged during cryostat sectioning.

  • Animals with injury severity that differs 2 SD from the mean as defined by the size of the injury site, which has to be within 2 SDs from the average.

No animal replacement was needed to ensure that the power requirements were met.

Specific calculations are found here below:

Immunofluorescence (control vs. TTK21): change in fluorescence intensity of 1.5-fold (between 50 and 75 arbitrary units for example); SD = 10 and 12; P: 0.05; Power 90%; N = 4

Axonal regeneration (control vs. TTK21): increase in % of axons regenerating beyond the lesion site from 5 to 10; SD = 2 and 4; Power 90%; P: 0.05, N: 8

Behavioral tests (gridwalk as example test, control vs. TTK21): changes in score from 4 to 2; SD: 2 and 1; Power 90%; P: 0.05; N = 12

Results were expressed as mean ± SEM. Statistical analysis was carried out using GraphPad Prism 9. Normality was tested for using the Shapiro–Wilk test. Normally distributed data were evaluated using a two-tailed unpaired Student t test or a two-way repeated measures ANOVA when experiments contained more than 2 groups. The Tukey or Sidak post hoc test was applied when appropriate. The Mann–Whitney U test was used for nonparametric evaluation. Given the level of nonsignificance in behavioural experiments, a frequentist equivalence test using the two one-sided tests (TOST) rule was needed to strengthen the null hypothesis conclusion (TOSTER 0.4.1, parameters 0.18.1.6, R 4.1.2).

A threshold level of significance α was set at P value <0.05. Significance levels were defined as follows: * P value < 0.05; ** P value < 0.01; *** P value < 0.001, **** P value < 0.0001. All data analyses were performed blind to the experimental group.

URL to this deposited stage 1 manuscript: 10.17605/OSF.IO/S5EDH

Supporting information

S1 Fig

(A) Twelve weeks old mice were housed in SH or EE 1 week after a spinal cord transection. (B and C) Animals in SH remained impaired unable to step until day 42 after injury as shown by BMS (B) and Gridwalk (C). EE significantly enhanced locomotion (mean ± SEM, two-way ANOVA, Fisher LSD post hoc ** P < 0.01; *** P < 0.005; **** P < 0.001). The data can be found in S1 Data. BMS, Basso Mouse Scale; EE, enriched environment; SH, standard housing.

(PDF)

S2 Fig. Graphical diagram summarizing the experimental design. Made with BioRender.

(PNG)

S3 Fig

(A) Representative micrographs of cJUN immunostaining (green, white arrows) in layer 5 cortical neurons from CSP or CSP-TTK21-treated mice. (B) Quantification of cJUN immunostaining in layer 5 cortical neurons (CSP: 2,539.0 ± 295.4; CSP-TTK21: 2,355.0 ± 206.7, p = 0.63, TOST: t(0.1) = 0.1, p = 0.5 given equivalence bounds of −153.0 and 153.0 on a raw scale and an alpha of 0.05, n = 4). (C) Representative micrographs of SPRR1a immunostaining (green, white arrows) in layer 5 cortical neurons. (D) Quantification of SPRR1a immunostaining in layer 5 cortical neurons in CSP or CSP-TTK21 (CSP: 3,940.0 ± 491.6, n = 3; CSP-TTK21: 3,636.0 ± 350.3, n = 4; p = 0.08, TOST: t(4.8) = −1.9, p = 0.94 given equivalence bounds of −344.4 and 344.4 on a raw scale and an alpha of 0.05). Mean ± SEM; unpaired two-tailed Student t test or Welch t test. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; TOST, two one-sided tests.

(PNG)

S4 Fig

(A) Representative micrographs of GFAP intensity (red) around the SCI site (white asterisks) and cavity size (white dotted line) in CSP or CSP-TTK21 mice. (B) Quantification of cavity size in CSP or CSP-TTK21-treated mice (CSP: 471,574.0 ± 76,631.0, n = 8; CSP-TTK21: 486,466.0 ± 45,491.0, n = 14; p = 0.87; TOST: t(20.0) = 0.5, p = 0.31 given equivalence bounds of −56,344.2 and 56,344.2 on a raw scale and an alpha of 0.05). (C) Quantification of GFAP intensity in CSP or CSP-TTK21-treated mice (CSP: 1,272.0 ± 31.3, n = 3; CSP-TTK21: 1,339.0 ± 20.8, n = 4, p = 0.12, TOST: t(3.7) = −1.4, p = 0.88 given equivalence bounds of −14.5 and 14.5 on a raw scale and an alpha of 0.05). Mean ± SEM; unpaired two-tailed Student t test or Welch t test. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; SCI, spinal cord injury; TOST, two one-sided tests.

(PNG)

S5 Fig

(A) Representative micrographs of CD68 immunofluorescence (red) and DAPI (blue) around the SCI site (white asterisks) in CSP or CSP-TTK21-treated mice. Lesion site (white dotted line). (B) Quantification of CD68 intensity in CSP or CSP-TTK21-treated mice (CSP: 472.0 ± 8.1, n = 3 CSP-TTK21: 434.5 ± 37.8 n = 4, p = 0.44, TOST: t(3.3) = 0.5, p = 0.69 given equivalence bounds of −16.3 and 16.3 on a raw scale and an alpha of 0.05). Mean ± SEM; unpaired two-tailed Student t test. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; SCI, spinal cord injury; TOST, two one-sided tests.

(PNG)

S1 Data. Excel spreadsheets containing the quantitative data for each experiment as described in the results and figure legends.

(XLSX)

Abbreviations

BMS

Basso Mouse Scale

CBP

CREB-binding protein

CNS

central nervous system

CSP

carbon nanosphere

CST

corticospinal tract

DRG

dorsal root ganglion

EE

enriched environment

HAT

histone acetyltransferase

PCAF

p300/CBP-associated factor

RAG

regeneration-associated gene

SCI

spinal cord injury

SH

standard housing

TOST

two one-sided tests

Data Availability

All relevant data are within the paper and its Supporting Information files.

Funding Statement

ISRT translational award-P90397 to SDG Marina Romoli Onlus-P82836 to SDG Rosetrees Trust-P72986 to SDG Brain Research Trust-P73576 to SDG and J C Bose fellowship to TKK. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Di Giovanni S. Regeneration following spinal cord injury, from experimental models to humans: where are we? Expert Opin Ther Targets. 2006;10(3):363–76. doi: 10.1517/14728222.10.3.363 . [DOI] [PubMed] [Google Scholar]
  • 2.van Niekerk EA, Tuszynski MH, Lu P, Dulin JN. Molecular and Cellular Mechanisms of Axonal Regeneration After Spinal Cord Injury. Mol Cell Proteomics. 2016;15(2):394–408. Epub 2015/12/24. doi: 10.1074/mcp.R115.053751 ; PubMed Central PMCID: PMC4739663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Beauparlant J, van den Brand R, Barraud Q, Friedli L, Musienko P, Dietz V, et al. Undirected compensatory plasticity contributes to neuronal dysfunction after severe spinal cord injury. Brain. 2013;136:3347–61. doi: 10.1093/brain/awt204 WOS:000326291800018. [DOI] [PubMed] [Google Scholar]
  • 4.van den Brand R, Heutschi J, Barraud Q, DiGiovanna J, Bartholdi K, Huerlimann M, et al. Restoring Voluntary Control of Locomotion after Paralyzing Spinal Cord Injury. Science. 2012;336(6085):1182–5. doi: 10.1126/science.1217416 WOS:000304647900056. [DOI] [PubMed] [Google Scholar]
  • 5.Wagner FB, Mignardot JB, Le Goff-Mignardot CG, Demesmaeker R, Komi S, Capogrosso M, et al. Targeted neurotechnology restores walking in humans with spinal cord injury. Nature. 2018;563(7729):65–+. doi: 10.1038/s41586-018-0649-2 WOS:000448900900046. [DOI] [PubMed] [Google Scholar]
  • 6.Wang ZM, Mehra V, Simpson MT, Maunze B, Chakraborty A, Holan L, et al. KLF6 and STAT3 co-occupy regulatory DNA and functionally synergize to promote axon growth in CNS neurons. Sci Rep. 2018;8. doi: 10.1038/s41598-018-31101-5 WOS:000442387900029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Blackmore MG, Wang Z, Lerch JK, Motti D, Zhang YP, Shields CB, et al. Kruppel-like Factor 7 engineered for transcriptional activation promotes axon regeneration in the adult corticospinal tract. Proc Natl Acad Sci U S A. 2012;109(19):7517–22. Epub 2012/04/25. doi: 10.1073/pnas.1120684109 ; PubMed Central PMCID: PMC3358880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.De Virgiliis F, Hutson TH, Palmisano I, Amachree S, Miao J, Zhou LM, et al. Enriched conditioning expands the regenerative ability of sensory neurons after spinal cord injury via neuronal intrinsic redox signaling. Nat Commun. 2020;11(1). doi: 10.1038/s41467-020-20179-z WOS:000603278300004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mehta ST, Luo XT, Park KK, Bixby JL, Lemmon VP. Hyperactivated Stat3 boosts axon regeneration in the CNS. Exp Neurol. 2016;280:115–20. doi: 10.1016/j.expneurol.2016.03.004 WOS:000375822000013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wolfgang PT, Marcus M, Avni J, Di G, Valeria C. HDAC5 promotes optic nerve regeneration by activating the mTOR pathway. Exp Neurol. 2019;317:271–83. doi: 10.1016/j.expneurol.2019.03.011 WOS:000470803400025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Raivich G, Bohatschek M, Da Costa C, Iwata O, Galiano M, Hristova M, et al. The AP-1 transcription factor c-jun is required for efficient axonal regeneration. Neuron. 2004;43(1):57–67. doi: 10.1016/j.neuron.2004.06.005 WOS:000222549800007. [DOI] [PubMed] [Google Scholar]
  • 12.Gao Y, Deng KW, Hou JW, Bryson JB, Barco A, Nikulina E, et al. Activated CREB is sufficient to overcome inhibitors in myelin and promote spinal axon regeneration in vivo. Neuron. 2004;44(4):609–21. doi: 10.1016/j.neuron.2004.10.030 WOS:000225338100008. [DOI] [PubMed] [Google Scholar]
  • 13.Belin S, Nawabi H, Wang C, Tang SJ, Latremoliere A, Warren P, et al. Injury-Induced Decline of Intrinsic Regenerative Ability Revealed by Quantitative Proteomics. Neuron. 2015;86(4):1000–14. doi: 10.1016/j.neuron.2015.03.060 WOS:000354878400014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zou HY, Ho C, Wong KR, Tessier-Lavigne M. Axotomy-Induced Smad1 Activation Promotes Axonal Growth in Adult Sensory Neurons. J Neurosci. 2009;29(22):7116–23. doi: 10.1523/JNEUROSCI.5397-08.2009 WOS:000266632400003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Farrukh F, Davies E, Berry M, Logan A, Ahmed Z. BMP4/Smad1 Signalling Promotes Spinal Dorsal Column Axon Regeneration and Functional Recovery After Injury. Mol Neurobiol. 2019;56(10):6807–19. doi: 10.1007/s12035-019-1555-9 WOS:000486010800010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Finelli MJ, Wong JK, Zou HY. Epigenetic Regulation of Sensory Axon Regeneration after Spinal Cord Injury. J Neurosci. 2013;33(50):19664–76. doi: 10.1523/JNEUROSCI.0589-13.2013 WOS:000328626800026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gaub P, Joshi Y, Wuttke A, Naumann U, Schnichels S, Heiduschka P, et al. The histone acetyltransferase p300 promotes intrinsic axonal regeneration. Brain. 2011;134:2134–48. doi: 10.1093/brain/awr142 WOS:000292049600027. [DOI] [PubMed] [Google Scholar]
  • 18.Puttagunta R, Tedeschi A, Soria MG, Hervera A, Lindner R, Rathore KI, et al. PCAF-dependent epigenetic changes promote axonal regeneration in the central nervous system. Nat Commun. 2014;5. doi: 10.1038/ncomms4527 WOS:000209867600001. [DOI] [PubMed] [Google Scholar]
  • 19.Palmisano I, Danzi MC, Hutson TH, Zhou L, McLachlan E, Serger E, et al. 7 Epigenomic signatures underpin the axonal regenerative ability of dorsal root ganglia sensory neurons. Nat Neurosci. 2019;22(11):1913–1924. doi: 10.1038/s41593-019-0490-4 [DOI] [PubMed] [Google Scholar]
  • 20.Hervera A, Zhou L, Palmisano I, McLachlan E, Kong G, Hutson TH, et al. PP4-dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure. EMBO J. 2019;38(13):e101032. Epub 2019/07/04. doi: 10.15252/embj.2018101032 ; PubMed Central PMCID: PMC6600644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hutson TH, Kathe C, Palmisano I, Bartholdi K, Hervera A, De Virgiliis F, et al. Cbp-dependent histone acetylation mediates axon regeneration induced by environmental enrichment in rodent spinal cord injury models. Sci Transl Med. 2019;11(487). Epub 2019/04/12. doi: 10.1126/scitranslmed.aaw2064 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chatterjee S, Mizar P, Cassel R, Neidl R, Selvi BR, Mohankrishna DV, et al. A Novel Activator of CBP/p300 Acetyltransferases Promotes Neurogenesis and Extends Memory Duration in Adult Mice. J Neurosci. 2013;33(26):10698–712. doi: 10.1523/JNEUROSCI.5772-12.2013 WOS:000320928900015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chatterjee S, Cassel R, Schneider-Anthony A, Merienne K, Cosquer B, Tzeplaeff L, et al. Reinstating plasticity and memory in a tauopathy mouse model with an acetyltransferase activator. EMBO Mol Med. 2018;10(11). doi: 10.15252/emmm.201708587 WOS:000449833900012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Joshi Y, Soria MG, Quadrato G, Inak G, Zhou L, Hervera A, et al. The MDM4/MDM2-p53-IGF1 axis controls axonal regeneration, sprouting and functional recovery after CNS injury. Brain. 2015;138(Pt 7):1843–62. Epub 2015/05/20. doi: 10.1093/brain/awv125 . [DOI] [PubMed] [Google Scholar]
  • 25.Poplawski GHD, Kawaguchi R, Van Niekerk E, Lu P, Mehta N, Canete P, et al. Injured adult neurons regress to an embryonic transcriptional growth state. Nature. 2020;581(7806):77–82. doi: 10.1038/s41586-020-2200-5 [DOI] [PubMed] [Google Scholar]
  • 26.Sagner A, Briscoe J. Establishing neuronal diversity in the spinal cord: a time and a place. Development. 2019;146(22). Epub 20191125. doi: 10.1242/dev.182154 . [DOI] [PubMed] [Google Scholar]
  • 27.Kumamaru H, Lu P, Rosenzweig ES, Kadoya K, Tuszynski MH. Regenerating Corticospinal Axons Innervate Phenotypically Appropriate Neurons within Neural Stem Cell Grafts. Cell Rep. 2019;26(9):2329–39 e4. doi: 10.1016/j.celrep.2019.01.099 ; PubMed Central PMCID: PMC6487864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Freeman R, Han M, Álvarez Z, Lewis JA, Wester JR, Stephanopoulos N, et al. Reversible self-assembly of superstructured networks. Science. 2018;362(6416):808–813. doi: 10.1126/science.aat6141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Du K, Zheng S, Zhang Q, Li S, Gao X, Wang J, et al. Pten Deletion Promotes Regrowth of Corticospinal Tract Axons 1 Year after Spinal Cord Injury. J Neurosci. 2015;35(26):9754–63. doi: 10.1523/JNEUROSCI.3637-14.2015 ; PubMed Central PMCID: PMC6605149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cheng L, Sami A, Ghosh B, Goudsward HJ, Smith GM, Wright MC, et al. Respiratory axon regeneration in the chronically injured spinal cord. Neurobiol Dis. 2021;155:105389. Epub 20210508. doi: 10.1016/j.nbd.2021.105389 ; PubMed Central PMCID: PMC8197742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Joshi Y, Sória MG, Quadrato G, Inak G, Zhou L, Hervera A, et al. 36 The MDM4/MDM2-p53-IGF1 axis controls axonal regeneration, sprouting and functional recovery after CNS injury. Brain. 2015;138(7):1843–1862. doi: 10.1093/brain/awv125 [DOI] [PubMed] [Google Scholar]
  • 32.Floriddia EM, Rathore KI, Tedeschi A, Quadrato G, Wuttke A, Lueckmann JM, et al. p53 Regulates the Neuronal Intrinsic and Extrinsic Responses Affecting the Recovery of Motor Function following Spinal Cord Injury. J Neurosci. 2012;32(40):13956–70. Epub 2012/10/05. doi: 10.1523/JNEUROSCI.1925-12.2012 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Basso DM, Fisher LC, Anderson AJ, Jakeman LB, McTigue DM, Popovich PG. Basso mouse scale for locomotion detects differences in recovery after spinal cord in ury in five common mouse strains. J Neurotrauma. 2006;23(5):635–59. doi: 10.1089/neu.2006.23.635 WOS:000237529200004. [DOI] [PubMed] [Google Scholar]
  • 34.Steward O, Zheng BH, Tessier-Lavigne M. False resurrections: Distinguishing regenerated from spared Axons in the injured central nervous system. J Comp Neurol. 2003;459(1):1–8. doi: 10.1002/cne.10593 WOS:000181744600001. [DOI] [PubMed] [Google Scholar]
  • 35.Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8(6):e1000412. Epub 2010/07/09. doi: 10.1371/journal.pbio.1000412 ; PubMed Central PMCID: PMC2893951. [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Gabriel Gasque

1 Jun 2021

Dear Simone,

Thank you for submitting your manuscript entitled "Combinatorial small molecule-mediated activation of CBP/p300 with environmental enrichment in chronic severe experimental spinal cord injury to enable axon regeneration and sprouting for functional recovery" for consideration as a Preregistered Research Article by PLOS Biology. Please accept my apologies for the delay in sending the decision below to you.

Your manuscript has now been evaluated by the PLOS Biology editorial staff. We have also discussed your proposal with two academic editors, one with expertise in the biological questions you are addressing and another one with expertise in Pre-registered Reports. I am writing to let you know that we are interested in peer-reviewing your proposal, but before we can do that, we would like you to address some concerns raised by the Academic Editor with expertise in Pre-registered Reports. These issues are very likely to come up with the reviewers and so, we think, addressing them now will save you time in the end. You can find the comments from the Academic Editors below my signature. When you re-submit, please provide a point-by-point response to her/his concerns. The Academic Editor is willing to answer any question you might have during the revision process. I would also be happy to answer any questions, via e-mail or phone/zoom.

In addition, and BEFORE you start your revision, we need you to complete your submission by providing the metadata that is required for full assessment. To this end, please login to Editorial Manager where you will find the paper in the 'Submissions Needing Revisions' folder on your homepage. Please click 'Revise Submission' from the Action Links and complete all additional questions in the submission questionnaire. (YOU DONT NEED TO SUBMIT YOUR REVISION YET, JUST COMPLETE THE METADATA).

Please re-submit your manuscript with your metadata within two working days, i.e. by Jun 03 2021 11:59PM.

Login to Editorial Manager here: https://www.editorialmanager.com/pbiology

During resubmission, you will be invited to opt-in to posting your pre-review manuscript as a bioRxiv preprint. Visit http://journals.plos.org/plosbiology/s/preprints for full details. If you consent to posting your current manuscript as a preprint, please upload a single Preprint PDF when you re-submit.

Once your full submission is complete, your paper will undergo a series of checks. Once they are complete, I will stamp a Major Revision decision to give you time to address the concerns below.

Feel free to email us at plosbiology@plos.org or ggasque@plos.org if you have any queries relating to your submission.

Kind regards,

Gabriel Gasque

Senior Editor

PLOS Biology

ggasque@plos.org

============================

Academic Editor’s comments:

What is the source of the results presented in Supporting Figure 1? Is this pilot data or from a previous published article?

Please make clearer which outcomes would disconfirm the hypothesis, and for more complex hypotheses in which there are multiple measured variables, what strength of confirmation or disconfirmation would be associated with what combination of outcomes?

Please provide all G*Power outputs (e.g. screenshots of the output screen) as I couldn't reproduce all of these calculations.

On p17 the authors note that the “specific effect size has been estimated based upon similar studies showing significant differences between experimental and control group.” More detail is required here for a Stage 1 Registered Report. The studies and effect size estimates that furnish these estimates need to be sourced, e.g. in a table, listing the test and effect size reported in each previous study. Given that the target effect sizes are based on existing research, the authors should also be sure to take into account the effects of selection bias and publication bias, which typically inflate published effect size estimates? As note in the RR guidelines, since publication bias over-inflates published estimates of effect size, power analysis must be based on the *lowest* available or meaningful estimate of the effect size (i.e. the lower end of the effect size distribution).

For the hypotheses involving gridwalk and BMS data, the authors propose a repeated measures ANOVA, but it isn’t clear what factors are included in this analysis. The framing of the hypothesis in the design table instead suggests it would be tested through a pairwise group comparison. If there is a factor of group as well as a repeated measures factor, then presumably the authors instead intend to use a mixed ANOVA, not a repeated measures ANOVA.

To maximise clarity of the study procedure, I recommend including a schematic/figure that depicts the sequence of interventions and measurements for the two groups.

As I understand it from pp7-8, the control group will also be exposed to an enriched environment (EE) but will receive a control intervention (nanoparticles and vehicle alone). Given that the key comparison is (CSP-TTK21 + EE) vs (control treatment + EE), and EE is therefore held constant between the groups, how is it possible for the design to reveal any *combined* effects of CSP-TTK21 and EE, as opposed to testing the effects of CSP-TTK21 alone?

Update the design table to make clear the sample size that will be recruited based on the alpha level that will be used to conclude support for the hypothesis (e.g. do not report two sample sizes, for p<.05 for p<.01, as it is unclear which sample size will be included).

The authors note in the introduction that “lack of validation of this hypothesis will also provide essential information allowing the scientific community to entertain alternative hypotheses.” Since statistically non-significant results can only provide weak evidence for invariance between conditions (i.e. absence of evidence rather than evidence of absence), if the authors want to be able to draw stronger conclusions in the event of non-significant results, they may wish to consider frequentist equivalance tests (https://journals.sagepub.com/doi/10.1177/2515245918770963) or the use Bayes factors (https://www.frontiersin.org/articles/10.3389/fpsyg.2014.00781/full). Unlike conventional NHST, these tests can provide positive evidence of no effect.

One of the key criteria that reviewers are asked to assess in Stage 1 RRs is "Whether the authors have pre-specified sufficient outcome-neutral tests for ensuring that the results obtained are able to test the stated hypotheses, including positive controls and quality checks.", and successfully passing such tests is an editorial criterion at Stage 2 following completion of the study. Your protocol does not obviously propose any such tests - therefore please consider whether such positive controls or data quality checks are appropriate, and if possible how they might be included in the design. For instance, what positive control might be included (independently of the main hypotheses) to confirm that the CSP-TTK21 treatment was administered successfully? To consider what control is the most appropriate: imagine you ran the study and found null or confusing results. What positive control would convince a skeptic that the intervention was administered with sufficient precision and reliability to be able to provide a fair test of the hypothesis?

p12 mentions sham surgery, but it is unclear how this factors into the study design. My understanding from introduction was that *all* mice will receive the spinal cord injury?

On p17 the authors note that “two animals will be added to each experiment based upon the probability of losing animals due to the experimental procedure such as spinal surgery.” What happens if more animals are lost than anticipated? Will they be replaced until the minimum sample size is achieved, or there is a hard limit on the number of animals that can be tested?

Please ensure that exclusion criteria for data are comprehensively and precisely pre-specified as it usually not possible to adjust these for preregistered analyses after provisional acceptance is granted. At present the only reference to exclusion criteria is pp17-18: “Exclusion criteria include tissues with low quality for further experimentation or imaging as well as animals with injury severity beyond 2 SD from the mean.” This requires elaboration. How is “low quality” defined? Objectively or subjectively and at what point in the study timeline will this be assessed? How is injury severity defined? Why 2 SDs? Are excluded animals replaced to ensure the power requirements are met? Are there any other exclusion criteria of any kind? I suggest listing the exclusion criteria precisely in bullet point form and ensuring they are as exhaustive as possible.

Decision Letter 1

Gabriel Gasque

4 Jun 2021

Dear Simone,

Thank you very much for submitting your manuscript "Combinatorial small molecule-mediated activation of CBP/p300 with environmental enrichment in chronic severe experimental spinal cord injury to enable axon regeneration and sprouting for functional recovery" for consideration as a Preregistered Research Article at PLOS Biology. As mentioned previously, manuscript has been evaluated by the PLOS Biology editors and by two Academic Editors with relevant expertise. One to the Academic Editors has provided some detailed feedback that we think you should address before we can send your paper to independent reviewers.

We anticipate addressing these issues should not take you very long. Thus, we expect to receive your revised manuscript within 1 month. You can find the detailed comments below my signature.

Please email us (plosbiology@plos.org) if you have any questions or concerns, or would like to request an extension. At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not intend to submit a revision so that we may end consideration of the manuscript at PLOS Biology.

**IMPORTANT - SUBMITTING YOUR REVISION**

Your revisions should address the specific points made by the Academic Editor. Please submit the following files along with your revised manuscript:

1. A 'Response to Reviewers' file - this should detail your responses to the editorial requests, present a point-by-point response to all of the comments, and indicate the changes made to the manuscript.

*NOTE: In your point by point response to the reviewers, please provide the full context of each review. Do not selectively quote paragraphs or sentences to reply to. The entire set of reviewer comments should be present in full and each specific point should be responded to individually.

You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

2. In addition to a clean copy of the manuscript, please also upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Related" file type.

*Resubmission Checklist*

When you are ready to resubmit your revised manuscript, please refer to this resubmission checklist: https://plos.io/Biology_Checklist

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision:

*Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*PLOS Data Policy*

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5

*Blot and Gel Data Policy*

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Gabriel Gasque

Senior Editor

PLOS Biology

ggasque@plos.org

*****************************************************

Academic Editor's comments:

What is the source of the results presented in Supporting Figure 1? Is this pilot data or from a previous published article?

Please make clearer which outcomes would disconfirm the hypothesis, and for more complex hypotheses in which there are multiple measured variables, what strength of confirmation or disconfirmation would be associated with what combination of outcomes?

Please provide all G*Power outputs (e.g. screenshots of the output screen) as I couldn't reproduce all of these calculations.

On p17 the authors note that the “specific effect size has been estimated based upon similar studies showing significant differences between experimental and control group.” More detail is required here for a Stage 1 Registered Report. The studies and effect size estimates that furnish these estimates need to be sourced, e.g. in a table, listing the test and effect size reported in each previous study. Given that the target effect sizes are based on existing research, the authors should also be sure to take into account the effects of selection bias and publication bias, which typically inflate published effect size estimates? As note in the RR guidelines, since publication bias over-inflates published estimates of effect size, power analysis must be based on the *lowest* available or meaningful estimate of the effect size (i.e. the lower end of the effect size distribution).

For the hypotheses involving gridwalk and BMS data, the authors propose a repeated measures ANOVA, but it isn’t clear what factors are included in this analysis. The framing of the hypothesis in the design table instead suggests it would be tested through a pairwise group comparison. If there is a factor of group as well as a repeated measures factor, then presumably the authors instead intend to use a mixed ANOVA, not a repeated measures ANOVA.

To maximise clarity of the study procedure, I recommend including a schematic/figure that depicts the sequence of interventions and measurements for the two groups.

As I understand it from pp7-8, the control group will also be exposed to an enriched environment (EE) but will receive a control intervention (nanoparticles and vehicle alone). Given that the key comparison is (CSP-TTK21 + EE) vs (control treatment + EE), and EE is therefore held constant between the groups, how is it possible for the design to reveal any *combined* effects of CSP-TTK21 and EE, as opposed to testing the effects of CSP-TTK21 alone?

Update the design table to make clear the sample size that will be recruited based on the alpha level that will be used to conclude support for the hypothesis (e.g. do not report two sample sizes, for p<.05 for p<.01, as it is unclear which sample size will be included).

The authors note in the introduction that “lack of validation of this hypothesis will also provide essential information allowing the scientific community to entertain alternative hypotheses.” Since statistically non-significant results can only provide weak evidence for invariance between conditions (i.e. absence of evidence rather than evidence of absence), if the authors want to be able to draw stronger conclusions in the event of non-significant results, they may wish to consider frequentist equivalance tests (https://journals.sagepub.com/doi/10.1177/2515245918770963) or the use Bayes factors (https://www.frontiersin.org/articles/10.3389/fpsyg.2014.00781/full). Unlike conventional NHST, these tests can provide positive evidence of no effect.

One of the key criteria that reviewers are asked to assess in Stage 1 RRs is "Whether the authors have pre-specified sufficient outcome-neutral tests for ensuring that the results obtained are able to test the stated hypotheses, including positive controls and quality checks.", and successfully passing such tests is an editorial criterion at Stage 2 following completion of the study. Your protocol does not obviously propose any such tests - therefore please consider whether such positive controls or data quality checks are appropriate, and if possible how they might be included in the design. For instance, what positive control might be included (independently of the main hypotheses) to confirm that the CSP-TTK21 treatment was administered successfully? To consider what control is the most appropriate: imagine you ran the study and found null or confusing results. What positive control would convince a skeptic that the intervention was administered with sufficient precision and reliability to be able to provide a fair test of the hypothesis?

p12 mentions sham surgery, but it is unclear how this factors into the study design. My understanding from introduction was that *all* mice will receive the spinal cord injury?

On p17 the authors note that “two animals will be added to each experiment based upon the probability of losing animals due to the experimental procedure such as spinal surgery.” What happens if more animals are lost than anticipated? Will they be replaced until the minimum sample size is achieved, or there is a hard limit on the number of animals that can be tested?

Please ensure that exclusion criteria for data are comprehensively and precisely pre-specified as it usually not possible to adjust these for preregistered analyses after provisional acceptance is granted. At present the only reference to exclusion criteria is pp17-18: “Exclusion criteria include tissues with low quality for further experimentation or imaging as well as animals with injury severity beyond 2 SD from the mean.” This requires elaboration. How is “low quality” defined? Objectively or subjectively and at what point in the study timeline will this be assessed? How is injury severity defined? Why 2 SDs? Are excluded animals replaced to ensure the power requirements are met? Are there any other exclusion criteria of any kind? I suggest listing the exclusion criteria precisely in bullet point form and ensuring they are as exhaustive as possible.

Decision Letter 2

Gabriel Gasque

28 Jul 2021

Dear Simone,

Thank you for submitting your manuscript "Small molecule-mediated activation of CBP/p300 with environmental enrichment to enable axon regeneration and sprouting for functional recovery in chronic severe experimental spinal cord injury" for consideration as a Preregistered Research Article at PLOS Biology. Your manuscript has been evaluated by the PLOS Biology editors, by an Academic Editor with relevant expertise, and by three independent reviewers. You will note that reviewer 3 has revealed his identity.

In light of the reviews (below), we will not be able to accept the current version of the manuscript, but we would welcome re-submission of a much-revised version that takes into account the reviewers' comments. We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is also likely to be sent for further evaluation by the reviewers.

We expect to receive your revised manuscript within 3 months.

Please email us (plosbiology@plos.org) if you have any questions or concerns, or would like to request an extension. At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not intend to submit a revision so that we may end consideration of the manuscript at PLOS Biology.

**IMPORTANT - SUBMITTING YOUR REVISION**

Your revisions should address the specific points made by each reviewer. Having discussed these comments with the academic editors, we would like you to consider the following:

1) Regarding reviewer 3's recommendation to use rats instead of mice, we do not think that it is necessary that you switch your model system as recommended. However, you should be reserved in your discussion of the true clinical impact of your work.

2) We agree with reviewer 3 that four groups should be tested for this study, even though previous studies have completed two of the groups.

3) We also think that negative results may be difficult to interpret. Thus, we would recommend that you are reserved in the conclusions you draw from the study given that hierarchy of synergy is not thoroughly tested. You may be able to address this with a clearer rationale of the treatment paradigm, as was requested by all the reviewers, but see point (5) below.

4) We agree with the concerns of the reviewers regarding positive controls and sample size, which go to the heart of the criteria for accepting a Registered Research Articles Stage 1. It will be important for you to thoroughly address these points (among the many others).

5) We would also like to see a more comprehensive specification of the proposed frequentist equivalence tests and/or Bayesian hypothesis tests (including, especially, the choice of prior for the Bayesian tests and justification of that prior) and the chosen parameters for the frequentist tests. You also need to be clear which tests, Bayesian or frequentist equivalence, will determine the interpretation in the event of negative results, as it is not guaranteed that they will produce equivalent strength of evidence.

Please submit the following files along with your revised manuscript:

1. A 'Response to Reviewers' file - this should detail your responses to the editorial requests (above), present a point-by-point response to all of the reviewers' comments, and indicate the changes made to the manuscript.

*NOTE: In your point by point response to the reviewers, please provide the full context of each review. Do not selectively quote paragraphs or sentences to reply to. The entire set of reviewer comments should be present in full and each specific point should be responded to individually, point by point.

You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

2. In addition to a clean copy of the manuscript, please also upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Related" file type.

*Re-submission Checklist*

When you are ready to resubmit your revised manuscript, please refer to this re-submission checklist: https://plos.io/Biology_Checklist

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision:

*Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*PLOS Data Policy*

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5

*Blot and Gel Data Policy*

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Gabriel Gasque

Senior Editor

PLOS Biology

ggasque@plos.org

*****************************************************

REVIEWS:

Reviewer #1: This study proposes to investigate whether the epigenetic strategies to stimulate regenerative gene expression program combined with neuronal activity-dependent enhancement of neuroplasticity and guidance can overcome the current inability to promote neurological recovery in severe and chronic spinal cord injury. Specially, the authors propose to deliver the small molecule CBP/p300 activator CSP-TTK21 in mice housed in an enriched environment.

The plan is to administer the CBP/p300 activator CSP-TTK21 (i.p.) once a week between week twelve and twenty following a severe transection model of spinal cord injury in the mouse. A control group of mice will be treated with nanoparticles and vehicle alone. The CSP-TTK21 and control nanoparticles will be used at the dosage of 20mg/kg that showed efficacy in subacute spinal cord injury as published recently.

Data analysis will assess modifications in regenerative signalling, in motor and sensory axon sprouting and regeneration, in synaptic plasticity as well as in neurological sensorimotor recovery.

Specifically, sprouting and regeneration of the dorsal columns will be analyzed with the retrograde axonal tracer Dextran (injected in the sciatic nerve in proximity of L4-L6 DRGs 7 days before sacrificing the animals). Sprouting and regeneration of the corticospinal

tracts (CSTs) will be analysed with stereotaxic injections of the neural tracer BDA into the

motor cortex 2 weeks before sacrifice. The authors will measure sprouting and regeneration of serotoninergic raphe-spinal motor tracts with 5-HT immunohistochemistry. To assess whether the CSP-TTK21 treatment will enhance synaptic plasticity, we will measure the number of inhibitory VGAT or excitatory VGLUT1/2 synaptic terminals in proximity of neuronal targets

such as interneurons in the dorsal horns and motoneurons in the ventral horns of the spinal cord (ChAT or NeuN immunostaining) including in association with specific tracing of CST,

sensory or 5-HT fibres.

Histone acetylation as read out of CBP/p300 activation will also be evaluated in layer V neurons, raphe nuclei and DRG neurons by immunofluorescence. The expression of several regeneration associated genes including ATF3, JUN, GAP43, SPRR1a, KLF family members, and STAT3 will also be studied by immunofluorescence in sensory and motor neurons.

The authors will assess locomotion, coordination and sensorimotor integration by performing

open field assessment with the BMS and the gridwalk tests. In addition, Von Frey test for mechanoception and mechanical allodynia as well as Hargreaves test for thermoception and thermal hyperalgesia will be used to specifically assess the function of the ascending sensory tracts.

Previously, the authors have reported that the small molecule proposed here was able to promote sensory axon regeneration and recovery after a dorsal hemisection SCI in mice. In that study, injured mice received a weekly intraperitoneal injection of CSP-TTK21 (20 mg/kg) or control CSP, beginning 4 hours after injury. The current study seeks to address whether delayed treatment in chronically injured animals can also promote axon regeneration, sprouting and functional recovery. Considering the unmet need to ameliorate and restore functions in chronic SCI patients, this study addresses important aspect of SCI. The results obtained from the proposed experiment may provide insightful clues about developing potential reparative therapy, determine whether growth promoting strategies and enhancement of neural activity are an effective therapeutic strategy in chronic spinal cord injury.

The proposed experiments are technically sound and feasible. Methods and reagents to be used are described clearly with relevant citations. Statistical analyses to be performed seem appropriate. Exclusion criteria for animals is adequately described. The authors have significant experience and have published several manuscripts in the past with similar study design.

There are only a few minor suggestions which could be addressed to further clarify the study.

-The authors had previously proposed that EE-induction of lasting increase in regeneration potential is mediated by a Cbp-dependent increase in histone acetylation and increase in gene expression, including pathways involved in neuronal activity, axonal projection and cytoskeleton remodeling. While EE likely affects multiple pathways and mechanisms that might act positively to improve SCI outcomes, the authors indicated that Cbp-dependent mechanisms likely mediate EE effects. As such, it is less clear why a strong synergy is expected when EE and the drug are combined.

-The sex of mice that will be used in this study is not mentioned.

-Expected timeline for the completion of the study was not included but would be helpful.

-The author might want to consider using Minimum Information about a Spinal Cord Injury Experiment (PMID: 24870067).

Reviewer #2: The proposed study takes an innovative and novel approach to a problem of very high importance. Coaxing repair from the central nervous system after injury is a long-standing challenge, and most work has focused on so-called acute interventions, which are applied immediately after injury. This fails to address the needs of millions of individuals with existing injuries, and even for future injuries the medical reality is that acute treatment is not always possible. The proposal here is to combine an enriched environment with a pharmacological treatment that targets an epigenetic mechanism, which will be administered many weeks after a spinal injury. A comprehensive battery of outcome measurements will then determine whether this treatment improves axon growth, animal behavior, and/or synapse formation in the damaged spinal cord.

The methodology and plans for analysis are relatively standard for the field and are well described, which raises confidence that the study will yield useful data (positive or negative) as planned.

There are some conceptual and technical issues that arise. I emphasize again the potential importance and novelty of this study; the comments below are offered in the spirit of helpful refinement or simply providing the authors a chance to help me better understand the rationale for some details.

1. The largest conceptual question: what is the rationale for supplying enriched environments one week after injury, but delayed CBP stimulation? And the closely related question: if the model is that EE is acting through a CBP mechanism, what is the rationale for providing EE to all animals and then additional CBP to half? The concern is that if EE already maximally engages the CBP-dependent pathway, there will be no added benefit to the pharmacological treatment. In general there is potential confusion here about the relationship between the mechanisms of the two treatments, how that relationship affects the rationale of their dual use, and how that relationship affects the selection of their timing.

2. Why is the CST being traced and analyzed, as opposed to the Gi as previously? The Gi is likely more relevant to locomotion, and there is already data to support some effect. The jump to CST is more likely to yield negative results and less likely to be related to the behaviors in question.

3. The study seems to lack positive controls. Given the extreme challenge of promoting axon growth in the chronic injury condition, negative results are quite possible. To plan for this, it seems important to have positive controls in place, especially for the histone and gene expression measurements.

4. A smaller technical question - why BDA for tracing CST axons? Viral tracing methods, used previously by this group, would seem to offer more sensitive detection of fine collaterals in the spinal grey matter. But perhaps this is not the case, or there may be other technical considerations that led to the selection of BDA?

Reviewer #3, Mark H. Tuszynski: This is an interesting proposal to study a combination of a slow-release nanoparticle to activate CREB-binding protein (CSP-TTK21) and environmental enrichment (EE) in mice after spinal cord injury (SCI). The work is proposed by a careful group that publishes high quality, well-documented studies in high impact journals.

The Di Giovanni group has previously published that EE enhances sensory axonal growth and motor behavioral recovery after SCI: EE increases H4K8ac protein levels and phosphorylation of CREB (Hutson TH et al, Sci Transl Med, 2019), as shown in Supporting Fig 1.

I have one major comment regarding the experimental design, several moderate comments, and some minor comments.

Major: Experimental groups - there will only be two experimental groups even though 2 therapies will be applied. I believe that for the emerging data from this study to be clearest and of the highest quality, there should be 4 groups: 1) Untreated control lesioned. 2) Treatment with EE. 3) Treatment with TTK21. 4) EE + TTK21. The authors may consider their previous report regarding the benefit of EE alone to be sufficiently compelling that they need not show the value of this alone, but I do not agree. This is a different study and there may be different effect sizes of EE, and synergies between EE and TTK21 that are only evident by examining adequate controls for each treatment.

Moderate:

Sampling Plan: The authors will study only 6 animals per group based on a power calculation that assumes a 200% increase in axon growth between the control and treatment group. This is very optimistic! I strongly suggest that the authors consider increasing the N per group to 12.

The functional studies also propose an effect size of 100%, which once again is quite optimistic (notwithstanding the preliminary data in Supp Fig 1). Again, I would increase group size to 12.

The authors propose to use the mouse model. Why use this small animal species as opposed to the rat? Data in rats may be more clinically relevant, and the authors state that they wish to design a clinically relevant experiment.

The authors reference "chronic" injury at several points of the paper, but EE is initiated one week after the injury in the proposed studies, not late, and a 3 month post-injury time point is not in any case necessarily "chronic". The authors might consider relying less heavily on this term. Perhaps more importantly, the authors might consider starting BOTH therapies 3 months after injury, or at least starting EE one month after injury: the average time in the U.S. that most patients enter rehabilitation is one month after injury, and few begin rehab one week after injury. But if the authors truly wish to study a late time point after injury when immediate post-injury events have subsided, initiating both therapies at 3 months would be better, in my opinion.

Minor:

The authors state that they will use a "severe" transection model of SCI, but a dorsal over hemisection is not that severe. Mouse with this injury spontaneously recover to a BMS score of nearly 3, are show a gradual improvement in function over 42 days, although the authors state that this model causes "permanent" deficits. These lesions spare the ventral motor tracts of the spinal cord (esp reticulospinal) which can support several motor behaviors. These statements do not entirely correspond to the presented data in Supporting Figure 1, and some clarification is needed both with regard to exactly what the lesion is, and why it is considered permanent.

The authors propose to use BDA as a tracer of the corticospinal system. Generally, our experience has been that newer fluorescent tracers such as TdTomato or membrane-trafficked tracers expressing fluorophores provide superior corticospinal axon visualization. The authors might consider using these tracers.

The authors propose to perform immunolabeling for a number of RAGs to gauge expression levels: this is some variability in these immunolabeling methods. Might it be possible to pursue a less extensive list of RAGs and study levels by, e.g., Western blot of quantitative in situ hybridization?

The authors inconsistently state that they will trace central projections of DRG axons with CTB in one part of the description, and Dextran red in another.

The authors provide supporting data in support of EE enhancing motor recovery. However, they do not provide anatomical data that supports their claim of axonal/synaptic reorganization. According to the text, this should be included in supporting figure 1.

For 5-HT immunolabelling, it is probably unnecessary to post-fix in glut, then perform antigen retrieval. In our experience, anti-5-HT antibodies (from Immunostar) perform well with 4% pfa fix without antigen retrieval.

Behavioral analysis: Assessments will be performed on week 1 post-injury and weekly thereafter, and CSP-TTK21 injections will be performed on the same day. I suggest that the motor assessments be performed prior to the IP injections.

Interpretations of negative outcomes: The authors very nicely propose several possible interpretations of negative outcomes (i.e., a lack of anatomical, behavioral or molecular differences from controls). But since this is a combinatorial treatment paradigm, there may in fact be many additional reasons that the approach could yield data. While all possibilities for a negative outcome are likely to be elucidated in this study, it remains worthy of study.

Overall, this is an interesting study that appears likely to yield useful and potentially important data. I hope that the preceding suggestions are helpful.

Decision Letter 3

Gabriel Gasque

14 Sep 2021

Dear Simone,

Thank you for submitting your revised Preregistered Research Article entitled "Small molecule-mediated activation of CBP/p300 with environmental enrichment to enable axon regeneration and sprouting for functional recovery in chronic severe experimental spinal cord injury" for publication in PLOS Biology. I have now obtained advice from the original reviewers and have discussed their comments with the Academic Editor. 

Based on the reviews, we are prepared to accept this proposal and to invite you to move ahead and do the experiments. However, before we do that, we would like to give you the opportunity to consider and respond, if you wish, to the lingering concerns expressed by reviewer 2. Addressing these concerns is not a requirement for eventual acceptance.

Please also include within your manuscript the ID number of the protocol approved by the Imperial College London Animal Welfare and Ethical Review Body Standing Committee (AWERB).

As you address these items, please take this last chance to review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the cover letter that accompanies your revised manuscript.

We expect to receive your revised manuscript within two weeks.

To submit your revision, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' to find your submission record. Your revised submission must include the following:

-  a cover letter that should detail your responses to any editorial requests, if applicable, and whether changes have been made to the reference list

-  a Response to Reviewers file that provides a detailed response to the reviewers' comments (if applicable)

-  a track-changes file indicating any changes that you have made to the manuscript. 

NOTE: If Supporting Information files are included with your article, note that these are not copyedited and will be published as they are submitted. Please ensure that these files are legible and of high quality (at least 300 dpi) in an easily accessible file format. For this reason, please be aware that any references listed in an SI file will not be indexed. For more information, see our Supporting Information guidelines:

https://journals.plos.org/plosbiology/s/supporting-information  

*Published Peer Review History*

Please note that you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

Please do not hesitate to contact me should you have any questions.

Sincerely,

Gabriel Gasque, Ph.D.,

Senior Editor,

ggasque@plos.org,

PLOS Biology

------------------------------------------------------------------------

Reviewer remarks:

Reviewer #1: The revision has addressed sufficiently the concerns and questions raised by this reviewer.

Reviewer #2: The authors have modified and significantly strengthened the manuscript to address the main concerns raised in the prior round of review. Most of the points were convincingly addressed. The rationale for providing EE to all animals as a surrogate for the normally "enriched" environment that is available to human patients - or put another way, to undo the artificial deprivation that likely distorts results in many lab animals - is clear. Clarifying the technical rationale for focusing here on CST growth, and adding the possibility of tracing reticulospinal axons if possible, is a strong response. In the same way, the plans for axon tracing were clarified and strengthened.

A few lingering concerns exist but may be minor. One could quibble and point out that the rationale offered for combining EE and CBP stimulation somewhat missed the question. The authors have clarified that EE does not act solely through CBP; that is quite clear and was not the source of confusion. The question was the opposite - are we sure that CBP activation via CSP-TTK21 is offering a benefit beyond the stimulation of CBP already achieved through EE? In other words, is the pharmacological effect on CBP somehow stronger or broader than EE's effect on CBP? If not, the drug is simply duplicating an effect already present in EE animals. But on reflection, a stronger effect from the drug seems likely enough that the overall experiment is interesting and very worthwhile. I clarify this point only to sensitize the authors to the likelihood that other readers may wonder the same thing - not just whether EE is doing more than just activating CBP, which is now clarified in the revised manuscript, but also whether CSP-TTK21 is doing more to CBP than EE alone, which does not seem to be addressed. It would seem that both must be true to predict synergy.

Finally, in response to questions about positive controls for histone and gene expression outcomes, the authors reiterated the intention to measure histone acetylation, pointing to prior findings that CSP-TTK21 treatment increased H4K8ac. Again, this was understood when the question was raised. This prior finding was obtained by acute delivery, not in the chronic state, so even in a narrow sense it is not clear that this readout qualifies as a positive control per se; it seems more of an open question, whether the histones of chronically injured neurons will be modified in the same way. More broadly, it leaves unanswered any question about positive controls for the RAGs to be measured. In reading responses to other reviewers, it appears that the intention is to compare the immunofluorescence obtained in these samples to other datasets obtained previously in the lab. In light of the outstanding and extensive track record of the lab, this is probably acceptable, although of course it must be done cautiously.

Reviewer #3, M Tuszynski: The authors have responded well to my comments. No additional suggestions.

Decision Letter 4

Gabriel Gasque

17 Sep 2021

Dear Simone,

Thank you for submitting your revised manuscript "Small molecule-mediated activation of CBP/p300 with environmental enrichment to enable axon regeneration and sprouting for functional recovery in chronic severe experimental spinal cord injury" for consideration as a Preregistered Research Article at PLOS Biology. Your Stage 1 manuscript has now been evaluated by the PLOS Biology editors, who have determined that your Stage 1 Protocol meets our criteria for importance of research question and technical soundness of the study proposal. We would therefore like to invite you to complete the study, as proposed, and submit the Stage 2 manuscript. Please carefully read all the following information.

*Explanation of Decision*

This is a Stage 1 'in-principle acceptance' decision, with a commitment to publish the final Stage 2 Preregistered Research Article (after revision, if needed), pending successful completion of the study according to these Stage 1 approved methods and analytic procedures, as well as an evidence-based interpretation of the results. Please see here for review criteria for Stage 2 manuscripts:

https://journals.plos.org/plosbiology/s/reviewer-guidelines#loc-reviewing-preregistered-research-articles

Editorial decisions will not subsequently be based on the perceived importance or novelty of the results obtained during the Stage 2 study. It is critical however that you adhere exactly to this approved Stage 1 study design when performing the study. Any deviation from these experimental procedures could lead to rejection of the manuscript at Stage 2. Please consult the editors immediately for advice if you need to alter this approved study plan.

**IMPORTANT**: Please follow the link below for important information regarding the Stage 2 manuscript template and review criteria. Please carefully read the guidelines on Stage 2 data collection BEFORE performing your study and completing your Stage 2 manuscript.

AUTHOR GUIDELINES: https://plos.io/AuthorGuidelines

*Depositing this Stage 1 Protocol*

PLOS Biology does not publish Stage 1 Protocols immediately following an in-principle acceptance. Instead they are held and integrated into a single, completed 'Preregistered Research Article' following review and acceptance of the final Stage 2 manuscript. You are however required to register this approved Stage 1 Protocol with the Center for Open Science (https://cos.io/prereg/) or another recognised repository. This may be done publicly or under private embargo until submission of the Stage 2 manuscript. Stage 1 Protocols can be quickly and easily registered using a tailored mechanism for Registered Reports (https://osf.io/rr/). Please do this now. You will need to include the URL to this deposited protocol in your Stage 2 manuscript.

*Timeline*

We understand that carrying out the study will require a significant length of time and and are willing to allow you six months to perform the study. Please email us at 'plosbiology@plos.org' to discuss this if you have any questions or concerns, or to discuss an alternate timeline.

At this stage, your manuscript remains formally under active consideration at our journal. Please notify us by email if you do not wish to submit a Stage 2 manuscript or wish to pursue publication elsewhere, so that we may end consideration of the manuscript at PLOS Biology.

*Resubmission Checklist*

Before submitting the Stage 2 manuscript, please review the following resubmission checklist: https://plos.io/Biology_Checklist

Please note that for PRA stage 2, the response to reviewers file does not follow the standard format, but should rather be a document for the reviewers detailing the changes made to the manuscript since the stage 1 accept.

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision:

*Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*PLOS Data Policy*

Please note that as a condition of publication, PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. Please note that for this article type, the raw data itself should be archived and made freely available in a public repository rather than submitted as supplementary material. Please make sure to read the Stage 2 submission guidelines online regarding how this data should be annotated and appropriately time stamped to show that data was collected after this Stage 1 in-principle acceptance and not before.

*Blot and Gel Data Policy*

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that, if applicable, you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosbiology/s/submission-guidelines#loc-materials-and-methods

Thank you again for your submission to PLOS Biology. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Gabriel Gasque

Senior Editor

PLOS Biology

ggasque@plos.org

Decision Letter 5

Kris Dickson

15 Jun 2022

Dear Simone,

It was very nice meeting you last week at the Keystone meeting. I'm following up with you on your Stage 2 PRA submission at PLOS Biology. I've discussed the submission with both of the academic editors we are consulting on this work (one is on the study as a technical expert, one as our resident PRA guru). The technical academic editor is happy to have this go back out for peer review. In order to make the path as smooth as possible, the PRA academic editor has, however, asked if you could address the following points before we send it back out for Stage 2 peer review. They've said:

1. As per the RR policy authors are required to register the approved Stage 1 protocol on the OSF at the point of IPA and include the URL to the Stage 1 manuscript in the Stage 2 manuscript. Please add this.

2. Reading the results as a non-specialist, I'm not seeing as clear a mapping between the analyses and the hypotheses as I would have expected. I suggest the following to make this clear: (a) add a column to the design table called "Outcome and conclusion" which briefly states whether the hypothesis was supported or unsupported. (b) in the results, make clear which analyses are preregistered and which are not, and whether the hypothesis in each case is supported. I suggest structuring the results around the research questions in the design table so that the mapping is absolutely clear.

3. Deviations from protocol (or at least wording)

* First paragraph of Experimental Design section (p7). There appears to be a significant change in phrasing here. Does this reflect a change in wording or a change in the actual procedure?

* Is there any significance in the different colours of tracked changes? The tracked changes at this point should show all changes from the approved Stage 1 manuscript and *only* those changes. I haven't had time to check the approved Stage 1 manuscript against this tracked changes version to see if this is the case (a staff editor should do this prior to review and ensure everything is sound)

* Animals were sacrificed at 22 weeks post-injury rather than 20 weeks as preregistered. Why was this? A Deviations from Protocol seciton should be added to the Methods that lists, explains and justifies any and all changes from the approved procedures, however minor / inconsequential that are. Note: this only applies to changes in procedures, i.e. what was actually done, not minor text alterations.

4. I didn't understand the rationale for not doing the frequentist equivalence tests (p23). The authors suggest that the level of nonsignificance was of a sufficient level not to require it, but this is not statistically coherent. Without equivalence tests, statements such as "CBP/p300 activator TTK21 does not affect neurological recovery in chronic severe SCI" are statistically unfounded because they rely on non-significant results (i.e. they are only absence of evidence, not evidence of absence). All preregistered tests need to be reported.

We expect to receive your revised manuscript within 1 month. Please email us (plosbiology@plos.org) if you have any questions or concerns, or would like to request an extension.

====

At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not intend to submit a revision so that we withdraw the manuscript.

**IMPORTANT - SUBMITTING YOUR REVISION**

Your revisions should address the specific points made by each reviewer. Please submit the following files along with your revised manuscript:

1. A 'Response to Reviewers' file - this should detail your responses to the editorial requests, present a point-by-point response to all of the reviewers' comments, and indicate the changes made to the manuscript.

*NOTE: In your point-by-point response to the reviewers, please provide the full context of each review. Do not selectively quote paragraphs or sentences to reply to. The entire set of reviewer comments should be present in full and each specific point should be responded to individually.

You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

2. In addition to a clean copy of the manuscript, please also upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Revised Article with Changes Highlighted " file type.

*Resubmission Checklist*

When you are ready to resubmit your revised manuscript, please refer to this resubmission checklist: https://plos.io/Biology_Checklist

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision:

*Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*PLOS Data Policy*

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5

*Blot and Gel Data Policy*

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Kris

Kris Dickson, Ph.D. (she/her)

Neurosciences Senior Editor/Section Manager

PLOS Biology

kdickson@plos.org

----------------------------------------------------------------

REVIEWS:

Decision Letter 6

Kris Dickson

5 Aug 2022

Dear Dr Di Giovanni,

Thank you for your patience while we considered your revised Stage 2 Preregistered Article "CBP/p300 activation promotes axon growth, sprouting and synaptic plasticity in chronic severe experimental spinal cord injury" for publication as a Preregistered Research Article at PLOS Biology. This revised version of your manuscript has been evaluated by the PLOS Biology editors, two Academic Editors (one topic-related; one to assess the Preregistration protocols) and the original reviewers.

Based on the reviews and our Academic Editor's assessment of your revision, we are likely to accept this manuscript for publication, provided you satisfactorily address the remaining points raised by the reviewers regarding the results and discussion. As this is a Preregistered Article (Registered Report), please disregard the comments from Reviewer 3 regarding suggested changes to the Introduction and Methods. Further, please ensure that the Introduction and Methods in the final revised manuscript match the prior approved Stage 1 Protocol manuscript. Textual changes should only be done to correct verb tense or, when necessary, to correct a factual error or to avoid a misunderstanding.

Please also provide a blurb which, if the paper is accepted, will be included in our weekly and monthly Electronic Table of Contents (eTOCs), sent out to readers of PLOS Biology. This blurb may also be used to promote your article on social media. The blurb should be about 30-40 words long and is subject to editorial changes. It should, without exaggeration, entice people to read your manuscript, should not be redundant with the title and should not contain acronyms or abbreviations. For examples, view our author guidelines: https://journals.plos.org/plosbiology/s/revising-your-manuscript#loc-blurb

Finally, please also make sure to address the data and other policy-related requests listed at the bottom of this email.

As you address these items, please take this last chance to review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the cover letter that accompanies your revised manuscript.

We expect to receive your revised manuscript within two weeks.

To submit your revision, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' to find your submission record. Your revised submission must include the following:

- a cover letter that should detail your responses to any editorial requests, if applicable, and whether changes have been made to the reference list

- a Response to Reviewers file that provides a detailed response to the reviewers' comments (if applicable)

- a track-changes file indicating any changes that you have made to the manuscript.

NOTE: If Supporting Information files are included with your article, note that these are not copyedited and will be published as they are submitted. Please ensure that these files are legible and of high quality (at least 300 dpi) in an easily accessible file format. For this reason, please be aware that any references listed in an SI file will not be indexed. For more information, see our Supporting Information guidelines:

https://journals.plos.org/plosbiology/s/supporting-information

*Published Peer Review History*

Please note that you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*Press*

Should you, your institution's press office or the journal office choose to press release your paper, please ensure you have opted out of Early Article Posting on the submission form. We ask that you notify us as soon as possible if you or your institution is planning to press release the article.

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Please do not hesitate to contact me should you have any questions.

Sincerely,

Kris

Kris Dickson, Ph.D. (she/her)

Neurosciences Senior Editor/Section Manager,

kdickson@plos.org,

PLOS Biology

------------------------------------------------------------------------

PREREGISTERED ARTICLE POLICIES:

In addition to the comments listed above regarding the introduction and methods sections, please move the public Stage 1 Protocol URL to the Methods section: https://osf.io/s5edh

More details on our guidelines for Preregistered Articles can be found here:

https://plos-marketing.s3.amazonaws.com/Marketing/Biology+Preregistered+Articles+Guidelines+for+Authors.pdf

------------------------------------------------------------------------

DATA POLICY:

You may be aware of the PLOS Data Policy, which requires that all data be made available without restriction: http://journals.plos.org/plosbiology/s/data-availability. For more information, please also see this editorial: http://dx.doi.org/10.1371/journal.pbio.1001797

Note that we do not require all raw data. Rather, we ask that all individual quantitative observations that underlie the data summarized in the figures and results of your paper be made available. We appreciate the provision of this data in the supplementary file you've provided.

We were not, however, able to locate the summary data for Supplemental Figure 1.

***Please add this data to the excel document.

***Please also ensure that figure legends in your manuscript include information on where the underlying data can be found, and ensure your supplemental data file/s has a legend.

Please ensure that your Data Statement in the submission system also accurately describes where your data can be found.

------------------------------------------------------------------------

DATA NOT SHOWN?

- Please note that per journal policy, we do not allow the mention of "data not shown", "personal communication", "manuscript in preparation" or other references to data that is not publicly available or contained within this manuscript. When going back over your study for final submission, please make sure to check for such statements, and either remove mention of any such data or provide figures presenting the results and the data underlying the figure(s).

------------------------------------------------------------------------

Reviewer remarks:

Reviewer's Responses to Questions

Do you want your identity to be public for this peer review?

Reviewer #1: No

Reviewer #2: No

Reviewer #3: Yes: Mark Tuszynski

Reviewer #1: In this study, the authors investigated whether delayed delivery of CBP/p300 activator TTK21 in adult mice after severe transection SCI in combination with enhanced environment (EE) housing promotes histone acetylation, axonal and synaptic plasticity and behavioral recovery.

Sprouting and regeneration of the dorsal columns and CSTs were analyzed with the retrograde and anterograde axonal tracers, respectively. Axonal dieback as well as the

number of fibres past the lesion site were normalized to the number of labelled fibres prior to

the lesion. The authors measured sprouting of serotoninergic raphe-spinal motor tracts with 5-HT immunohistochemistry. To assess whether the CSP-TTK21 treatment enhances synaptic

plasticity, they measured the number of inhibitory vGat or excitatory vGlut1 synaptic terminals

in proximity of neuronal targets such as interneurons in the dorsal horns and motoneurons in

the ventral horns of the spinal cord (ChAT or NeuN immunostaining).

Histone acetylation as read out of CBP/p300 activation was evaluated in layer V neurons, raphe nuclei and DRG neurons by immunofluorescence. The expression of several regeneration associated factors including ATF3, JUN, GAP43, SPRR1a, KLF7, pERK, and pSTAT3 was studied by immunofluorescence in sensory and motor neurons.

Locomotion, coordination and sensorimotor integration were measured by performing open field assessment with the BMS and the gridwalk tests. Von Frey test for mechanoception and mechanical allodynia as well as Hargreaves test for thermoception and thermal hyperalgesia were used to specifically assess the function of the ascending sensory tracts.

The authors conclude that TKK21 treatment promotes histone acetylation in both DRG and cortical neurons, increases expression of some RAGs in DRGs, prevents axonal dieback, promotes axonal sprouting, particularly for 5-HT axons. However, recovery of function was not observed in these TTK21-treated mice.

This study was set out to address an important question of whether previously identified regenerative treatment as shown in acute SCI models can promote plasticity and recovery in chronically injured animals. Importantly, the authors reason that EE does not represent a specific form of focused rehabilitation, but rather a more physiological setting compared to (standard housing) SH that better reflects the human condition where patients are encouraged to engage in physical activities after a spinal cord injury. There were several suggestions raised by the reviewers in the first round of review, most of which were decided not to be taken in the final study. These suggestions include inclusion of additional control groups, performing EE treatment months after SCI (i.e. at the time of TTK21 treatment) to reflect a delayed treatment paradigm, and choice of anterograde tracing method. However, it can be viewed that the authors present reasonable justification at least for the latter two recommendations.

From the study, the authors conclude that although TTK21 with EE promotes histone acetylation and expression of select RAGs in sensory neurons, this treatment paradigm alone is insufficient to promote recovery of functions. The authors discuss that the chronically injured environment of CNS and spinal cord might need further modifications to successfully induce functional recovery.

Th overall conclusion of the findings is an important addition to the SCI field and highlight the clear challenges for treating chronic SCI patients. The techniques used in this study are standard in the field and the authors have carried out carefully designed experiments to answer specific questions.

Minor points:

Figure 2A, there is a duplicate of NeuN image for the TTK21 group, and the ATF3 single channel image for the TTK21 group is missing.

Figure 3A-D, it is unclear if the rostral axons represent those labeled axons that have regenerated past the lesion site (i.e. distal to the lesion). If so, it is not easily seen these labelled axons in the representative images provided in Figure 3A and 3C.

The authors should consider citing these papers which seem relevant to the idea tested (i.e. challenges and possibilities in treating chronic SCI) and to the discussion of the findings. PMID: 26134657, PMID: 11717367, PMID: 33975016

Reviewer #2: The introduction, rationale and stated hypotheses are the same as the approved Stage 1 Protocol submission and experiments were executed and analyzed as planned.

I do have some questions about the presentation and conclusions presented in the abstract and discussion.

1. Overall there may be some tendency to advertise positive results but leave it to the reader to dig out the limitations and the effect size (which is quite small). The abstract in particular should give a more balanced and quantitative view of the data. The fact that RAGs were activated in DRG but not CST neurons is a significant finding. The fact that DRG and CST axon sprouting averaged less than 500 micros from the PROXIMAL edge of the lesion, and apparently didn't extend into tissue beyond the lesion, is certainly relevant information and informs the lack of behavioral effect. Bottom line, in my view the abstract should provide information about differences between the responses by different cell types and quantitative reference to the effect size of growth.

2. The injury is not sufficiently described, specifically the distinction between this injury, which is described as "severe" and a prior injury. This is important, because the difference in injury is offered as an explanation for the apparent reduction in DRG growth response in the prior acute study and the current chronic study.

From the present manuscript:

"A laminectomy at vertebra T9 was performed to expose spinal level T9 and a deep dorsal transection past the central canal leaving using micro-scissors (Fine Science Tools)."

From the prior manuscript (Joshi et al. 2015).

"A laminectomy at vertebra T9 was performed to expose spinal level T12 and a dorsal hemisection until the central canal was then performed using micro-scissors (Fine Science Tools)."

A more rigorous description of how depth was monitored, and how the depth differed between the two studies, is needed to support the claim that it can explain the reduced growth present here.

3. There seems to be a claim that TTK21 has different efficacy in sensory and motor neurons.

"It is however important that TTK21 increases the growth and regenerative gene expression ability of both sensory and motor neurons, albeit with differential potency and efficacy."

Is this referring to a difference in the response of CST and DRG in the present data? I don't see that in the present data or a statistical test to support that claim.

4. "These findings are in line with previous work in subacute SCI where these classical RAGs were not activated in the corticospinal as opposed to DRG neurons[27]. "

The reference is Dr. Tuszynski's claim that CST neurons temporarily revert to an embryonic state, and doesn't seem to support the claim being made for differential gene activation in DRG versus CST neurons.

Reviewer #3: Muller and colleagues report their followup study regarding CBP/p300 activation in a model of delayed SCI (T9 partial (?) transection 1and treatment 12 weeks after injury).

Abstract: The authors state: "The interruption of spinal circuitry following spinal cord injury disrupts neural activity AND IS FOLLOWED BY A FAILURE TO MOUNT AN EFFECTIVE REGENERATIVE GENE EXPRESSION RESPONSE resulting in permanent neurological disability." I believe that the statement in CAPS is not accurate. We reported in Nature 2020 that SCI DOES result in mounting regenerative gene expression, but the absence of a permissive milieu in the lesion site results in regenerative failure (Poplawski 2020; 581:77). I suggest that the authors simply state AND IS FOLLOWED BY A FAILURE TO MOUNT AN EFFECTIVE REGENERATIVE RESPONSE.

Related to the comment above, the authors may wish to reframe their conceptual context. We found as stated above that the regenerative state after SCI lasts for two weeks only. In a chronic state of injury (12-22 weeks after injury), this "primed" state for regeneration has closed. Thus, the authors may, by delivery of CSP-TTK, re-open this regenerative window. This concept is important from my perspective because we needs to identify means of re-opening the regenerative state.

Introduction: I suggest that you add PTEN and SOCS3 to the list genes that can influence regeneration, since the greatest evidence exists for PTEN. Although a phosphatase inhibitor, PTEN in effect acts like a transcription factor.

Hypothesis statement: The authors state the following: "Here we hypothesize that the pharmacological stimulation of CBP/p300 activity will enhance regenerative gene expression during a growth refractory phase twelve weeks after spinal injury, while housing animals in an EE one-week post-injury will stimulate neuronal activity, consolidate axonal and synaptic plasticity as opposed to animals housed in SH." I find this somewhat confusing and disorienting. Is this a chronic study (12 weeks after injury) or a sub-acute study (EE one week after injury)? I find the entire Hypothesis section difficult to follow; I encourage the authors to re-write it to enhance clarity and simplicity.

Experimental Design: This section states that anatomical transection in a mouse allows more accurate anatomical definition of regeneration and sprouting than contusion in rat. Please simply state that anatomical transection allows more accurate anatomical assessment of regeneration and sprouting, since the choice of mice or rats has no bearing on transection vs. contusion. Also the authors suggest that T9 transection may spare some axons. This is not the case is the lesions are accurately performed. The lesion images shown in the figures appear to spare a substantial amount of spinal cord tissue. I think this point needs clarification: either it is advisable to omit reference to "severe" SCI, or to clarify why the images that show a substantial amount of sparing in Fig 3 are "severe".

Regarding the finding that TTK increased axonal growth but not functional recovery: Yes, TTK significantly increased axon growth, but the effect size was not very large. One might think that this is the reason that function did not recover. Perhaps this should be stated simply and clearly. If one goes from no regenerating axons to a few, this may not be likely to exert much of a benefit on function.

Hypothesize is misspelled in the paper ("Hypothesise").

I greatly appreciate that the authors are reporting the fact that these experimental manipulations do not improve functional outcomes. Too often in this field, these negative findings are omitted from a paper. This complete reporting enhances the qualify and credibility of the work.

Decision Letter 7

Kris Dickson

12 Aug 2022

Dear Dr Di Giovanni,

Thank you for the submission of your revised Stage 2 Preregistered Research Article "CBP/p300 activation promotes axon growth, sprouting and synaptic plasticity in chronic experimental spinal cord injury with severe disability" for publication in PLOS Biology. On behalf of myself, my colleagues and the Academic Editors Cody Smith and Christopher Chambers, I am pleased to say that we can in principle accept your manuscript for publication, provided you address any remaining formatting and reporting issues. These will be detailed in an email you should receive within 2-3 business days from our colleagues in the journal operations team; no action is required from you until then. Please note that we will not be able to formally accept your manuscript and schedule it for publication until you have completed any requested changes.

Please take a minute to log into Editorial Manager at http://www.editorialmanager.com/pbiology/, click the "Update My Information" link at the top of the page, and update your user information to ensure an efficient production process.

PRESS

We frequently collaborate with press offices. If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximize its impact. If the press office is planning to promote your findings, we would be grateful if they could coordinate with biologypress@plos.org. If you have previously opted in to the early version process, we ask that you notify us immediately of any press plans so that we may opt out on your behalf.

We also ask that you take this opportunity to read our Embargo Policy regarding the discussion, promotion and media coverage of work that is yet to be published by PLOS. As your manuscript is not yet published, it is bound by the conditions of our Embargo Policy. Please be aware that this policy is in place both to ensure that any press coverage of your article is fully substantiated and to provide a direct link between such coverage and the published work. For full details of our Embargo Policy, please visit http://www.plos.org/about/media-inquiries/embargo-policy/.

Thank you again for choosing PLOS Biology for publication and supporting Open Access publishing. We look forward to publishing your study. 

Sincerely, 

Kris

Kris Dickson, Ph.D. (she/her)

Neurosciences Senior Editor/Section Manager

PLOS Biology

kdickson@plos.org

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig

    (A) Twelve weeks old mice were housed in SH or EE 1 week after a spinal cord transection. (B and C) Animals in SH remained impaired unable to step until day 42 after injury as shown by BMS (B) and Gridwalk (C). EE significantly enhanced locomotion (mean ± SEM, two-way ANOVA, Fisher LSD post hoc ** P < 0.01; *** P < 0.005; **** P < 0.001). The data can be found in S1 Data. BMS, Basso Mouse Scale; EE, enriched environment; SH, standard housing.

    (PDF)

    S2 Fig. Graphical diagram summarizing the experimental design. Made with BioRender.

    (PNG)

    S3 Fig

    (A) Representative micrographs of cJUN immunostaining (green, white arrows) in layer 5 cortical neurons from CSP or CSP-TTK21-treated mice. (B) Quantification of cJUN immunostaining in layer 5 cortical neurons (CSP: 2,539.0 ± 295.4; CSP-TTK21: 2,355.0 ± 206.7, p = 0.63, TOST: t(0.1) = 0.1, p = 0.5 given equivalence bounds of −153.0 and 153.0 on a raw scale and an alpha of 0.05, n = 4). (C) Representative micrographs of SPRR1a immunostaining (green, white arrows) in layer 5 cortical neurons. (D) Quantification of SPRR1a immunostaining in layer 5 cortical neurons in CSP or CSP-TTK21 (CSP: 3,940.0 ± 491.6, n = 3; CSP-TTK21: 3,636.0 ± 350.3, n = 4; p = 0.08, TOST: t(4.8) = −1.9, p = 0.94 given equivalence bounds of −344.4 and 344.4 on a raw scale and an alpha of 0.05). Mean ± SEM; unpaired two-tailed Student t test or Welch t test. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; TOST, two one-sided tests.

    (PNG)

    S4 Fig

    (A) Representative micrographs of GFAP intensity (red) around the SCI site (white asterisks) and cavity size (white dotted line) in CSP or CSP-TTK21 mice. (B) Quantification of cavity size in CSP or CSP-TTK21-treated mice (CSP: 471,574.0 ± 76,631.0, n = 8; CSP-TTK21: 486,466.0 ± 45,491.0, n = 14; p = 0.87; TOST: t(20.0) = 0.5, p = 0.31 given equivalence bounds of −56,344.2 and 56,344.2 on a raw scale and an alpha of 0.05). (C) Quantification of GFAP intensity in CSP or CSP-TTK21-treated mice (CSP: 1,272.0 ± 31.3, n = 3; CSP-TTK21: 1,339.0 ± 20.8, n = 4, p = 0.12, TOST: t(3.7) = −1.4, p = 0.88 given equivalence bounds of −14.5 and 14.5 on a raw scale and an alpha of 0.05). Mean ± SEM; unpaired two-tailed Student t test or Welch t test. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; SCI, spinal cord injury; TOST, two one-sided tests.

    (PNG)

    S5 Fig

    (A) Representative micrographs of CD68 immunofluorescence (red) and DAPI (blue) around the SCI site (white asterisks) in CSP or CSP-TTK21-treated mice. Lesion site (white dotted line). (B) Quantification of CD68 intensity in CSP or CSP-TTK21-treated mice (CSP: 472.0 ± 8.1, n = 3 CSP-TTK21: 434.5 ± 37.8 n = 4, p = 0.44, TOST: t(3.3) = 0.5, p = 0.69 given equivalence bounds of −16.3 and 16.3 on a raw scale and an alpha of 0.05). Mean ± SEM; unpaired two-tailed Student t test. n = biologically independent animals. The data can be found in S1 Data. CSP, carbon nanosphere; SCI, spinal cord injury; TOST, two one-sided tests.

    (PNG)

    S1 Data. Excel spreadsheets containing the quantitative data for each experiment as described in the results and figure legends.

    (XLSX)

    Attachment

    Submitted filename: Plos Biol response to reviewers 18-8-21 .docx

    Attachment

    Submitted filename: Mueller et al rebuttal Plos Biology revised 16-09-21.docx

    Attachment

    Submitted filename: Mueller et al rebuttal Plos Biology full sub.docx

    Attachment

    Submitted filename: Mueller et al rebuttal Plos Biology full sub.docx

    Attachment

    Submitted filename: Mueller et al Rebuttal Plos Biol minor rev 09-08-22.docx

    Data Availability Statement

    All relevant data are within the paper and its Supporting Information files.


    Articles from PLoS Biology are provided here courtesy of PLOS

    RESOURCES