ONLINE SUPPLEMENT

SUPPLEMENTARY METHODS

Drug storage and intranasal dosing. LM22A-4 was stored as a 10x solution (5 mg/ml in saline) at -80 °C. Aliquots were diluted with saline within 1 hour prior to administration. Each mouse was weighed weekly and the week’s dosage adjusted accordingly. For intranasal dosing, mice were manually restrained and inverted, and a P20 pipetman (Gilson) was used to place one drop at a time on one nostril until the liquid level fell beneath the external nares, and nostrils alternated each day of dosing.

Hypoxic-ischemic stroke. Mice were anesthetized using 2% isoflurane in 100% oxygen and body temperature was maintained at 37oC. To induce hypoxic-ischemic stroke the right common carotid artery was permanently ligated. Mice were then placed in a 37°C chamber containing 8% oxygen and 92% nitrogen for 25 minutes.

Western blots. Mice underwent hypoxic-ischemic stroke and those exhibiting >17% error on the horizontal ladder test were treated with daily LM22A-4 from days 3 to 10 after stroke, then sacrificed at 1 hour after the last dose of LM22A-4 or vehicle. Ipsilateral striatum was isolated, homogenized in cell lysis buffer, sonicated, and equalized as described.1 Protein extracts were subjected to Western blot analysis using standard techniques. Blots were probed with antibodies recognizing pTrkB-Y817 (rabbit mAb 1:30,000, Epitomics), TrkB (rabbit polyclonal antibody 1:2500, Millipore), and HRP goat anti-rabbit (1:10,000, Pierce Biotechnology). Optical densities were measured using UN-Scan-it Gel & Graph Digitizing Software Ver. 6.14. Ratios were calculated as pTrkB over full length TrkB receptor and data was normalized to saline-treated stroked mice. Mass spectroscopy. Mice underwent hypoxic-ischemic stroke and those exhibiting >17% error on the horizontal ladder test were treated with daily LM22A-4 from days 3 to 10 after stroke, then sacrificed at 1 hour after the last dose of LM22A-4 or vehicle. Ipsilateral hemispheres were obtained and immediately frozen on dry ice. The tissue extraction and LC-MS/MS analysis were performed by Absorption Systems (Exton, PA) as described.2 Brain homogenate from an untreated C57BL/6J mouse was used for the standard curve.

Behavioral tests. Prior to behavior training mice were handled three times, for 5 minutes each day. Behavioral apparatuses were cleaned in between mice with Simple Green. The same individual performed each behavioral test for the duration of each study, and this was never an individual who was also dosing mice. All individuals dosing and testing mice were blinded throughout the study, and dosing was performed after behavioral testing had been completed for the day.

Ladder test. The hypoxic-ischemic stroke model produces a deficit in this test in mice.3 A horizontal ladder was custom made from plexiglass and measured 30 inches across with rungs spaced 0.25 inches apart.4 It was supported on clean mouse cages and runs were filmed from underneath. To run each trial mice were removed from their home cage and placed at the end of 2 the ladder, next to a bright light. They then walked across the ladder back towards their home cage. Mice underwent four training sessions on the ladder in the four weeks prior to stroke. For the first session mice were sent across the ladder 3 times, then twice for the second training session, and only once for third and fourth training sessions. Baseline was obtained from the last training run. Post stroke testing was performed with one run per mouse. Ladder test performance was scored as the percent correct steps and the total number of left front foot missteps.

Automated gait analysis. Mice were trained on the automated gait analysis apparatus (Noldus Catwalk). This test was evaluated in a prior study (Pollak and Buckwalter, unpublished data), and limb swing speed and stride length were pre-specified variables for this study based on their high correlation with stroke size in the prior study. Mice were trained once a week for three weeks prior to stroke, and then tested weekly on day 5, 12, 19, 26, and 33 after stroke. For analysis, each mouse had to complete the run within 8 seconds and have at least 4 consecutive strides without pausing. Runs where the animal went backwards, reared, or paused excessively were disregarded. Two successful runs per mouse were obtained for each session.

Rotarod. This test has been previously used to follow deficits in the hypoxic-ischemic stroke model.5 Pre-stroke training was performed three times a week for four weeks prior to stroke on a mouse rotarod (Rotor-RodTM, San Diego Instruments). For training day 1 each mouse was placed on the stationary rotarod for 1 minute to allow it to get acclimated to the rotarod and the testing environment. For training day 2 the rotarod was turned on and set to run at a constant 5 rpm. Each mouse was placed on the moving rotarod until it could stay on the rotating rotarod for 1 minute continuously. For subsequent training and testing days the rotarod was set to accelerate from 0 to 5 rpm in the first ten seconds, then to 10 rpm over the next 290 seconds. Each mouse performed the test twice in a row and then the cohort was cycled through and the test repeated, for a total four trials per training or testing day. The length of time each mouse was able to stay on the rotarod, up to 300 seconds was recorded. Post stroke rotarod testing was performed weekly for 10 weeks after stroke, beginning on day 2 after stroke.

Stratification. Stratification was performed by one individual (MB) who assigned the stroked mice into two groups, A and B, that had day 1 ladder scores that were evenly distributed between groups, using the rotarod results from day 2 as a tiebreaker where needed. Sham mice were also assigned to groups A and B. A second individual (TY) prepared the drug and, without knowing their functional test results, assigned groups A and B to either LM22A-4 or saline. Dosing was then performed by individuals (JH, KPD, KTL, JZG, EC) who did not know whether A or B was the drug or the placebo control.

Perfusion and brain processing. Mice were sedated with 3.8% chloral hydrate perfused with 0.9% heparinized saline (10U/mL). The ipsilateral hemisphere was processed in 4% paraformaldehyde (PFA) in phosphate buffer, while the contralateral hemisphere was processed for Golgi-Cox impregnation. Ipsilateral hemispheres were fixed in 4% PFA for 24hr, rinsed with PBS, and sunk in 30% sucrose in PBS. Coronal brain sections 40µM thick were cut with a freezing sliding microtome (Microm HM430) into 24 sequential tubes, so that each tube contained every 24th section, and stored in cryoprotective medium at -20°C. The contralateral hemispheres were impregnated with Modified Golgi-Cox Staining Solution for 8 days at room temperature in the dark. The brains were then rinsed twice with dH2O and then transferred to 3

30% sucrose in dH2O for 3 days at 4˚C, with the solutions changed after the first initial 12 hours. The brains were then sectioned coronally using a vibratome (Leica VT10005) at 150µM in 30% sucrose in dH2O and mounted in 0.3% gelatin in dH2O. Once the gelatin solidified, the slide was immersed in 40% sucrose in dH2O and allowed to dry for 72hrs in the dark. Slides were then rinsed three times in dH2O for ten minutes, immersed in Developing Solution in for 7 minutes, and rinsed three times in dH2O for ten minutes each. The slides were then dehydrated through graded ethanol, followed by Histoclear (HS-200, National Diagnostics) and immediately cover- slipped with DPX mounting medium (EMS).

Immunohistochemistry, Hemisphere size quantification, and stereology. Immunohistochemistry was performed on PFA fixed, free-floating coronal brain sections (40µm) using standard techniques. All analysis was performed in a blinded fashion. The following primary antibodies were used: anti-NeuN (1:1000, Cat# MAB377, Millipore), anti-BrdU (1:5000, Cat# AB6326, Abcam), biotinylated anti-BrdU (1:500, Cat# AB2284, Abcam), anti- doublecortin (1:500; Cat# SC8066, Santa Cruz Biotechnology); anti-GFAP (1:1000, Cat# Z0334, DakoCytomation), anti-MHC II (1:500, Cat# 553621, BD Pharmingen), anti-CD68 (1:1000, Cat#MCA1957S, Serotec); anti-PECAM1/CD31 (1:300, Cat# 550274, BD Pharmingen). Hemisphere size was determined by tracing the remaining hemisphere size in every 24th section, spaced 960 µm apart, stained with Cresyl Violet. Stereological estimation of total BrdU+ cells was done on coronal sections spaced 480 µm apart and spanning the entire cortex and striatum (Stereo Investigator, MBF Bioscience). To determine the percentage of double-positive NeuN+/BrdU+ and PECAM+/BrdU+, the same areas were quantified in two sections spaced 960µm apart. From each section, 100 BrdU cells in each of the penumbral cortex, dorsolateral and ventral striatum were analyzed (40x objective). For doublecortin, the percent area covered in the entire section was measured in every 12th section spanning the entire brain. For GFAP, MHC II, and CD68, we quantified percent area covered in one 10x field of the penumbral cortex. For PECAM, we quantified percent area covered in one 10x field of the penumbral cortex from 3-5 coronal sections spaced 960 µm apart.

Analysis of Golgi stained neurons. Morphological reconstruction and analysis of pyramidal neurons in cortical layers 5/6 and 2/3, and medium spiny neurons in the dorsolateral striatum were performed using Neurolucida software (MBF Biosciences). Five neurons per region per mouse were analyzed. Dendritic spines were identified as protrusions along the dendrite axis and were traced at 100x objective. The spine densities of five secondary dendrites from layer 2/3 pyramidal neurons were analyzed using NeuroExplorer software (MBF Biosciences). Axonal Sprouting. Mice underwent dMCAO surgery at 5 months of age and were injected with 300nL 10% solution of biotinylated dextran amine (BDA; 10,000MW, Invitrogen) into the barrel cortex (-1.0mm bregma, 3.5.mm lateral) two weeks later. They were dosed with 0.22mg/kg/day LM22A-4 from day 3 to 21 after stroke. To measure axonal sprouting, 4 coronal sections that were 160 µm apart and centered on the injection core were used per animal. Streptavodin-488 (1:200, Cat#86493, Jackson ImmunoResearch) was used to detect BDA-labeled axons. Using a 40x objective, confocal stacks were taken in four areas of interest: contralesional dorsal striatum, ipsilesional dorsal striatum, penumbra, and the corpus callosum at midline. 4

SUPPLEMENTARY FIGURES AND FIGURE LEGENDS

Supplementary Figure 1. Histopathological features of hypoxic-ischemic stroke. Photomicrographs of Cresyl violet stained sequential coronal sections spaced 960 µm apart from two mice with typical lesion characteristics, sacrificed 6 weeks after stroke. Four sections are shown from each mouse. Injury occurred in large areas of cortex, hippocampus, and striatum ipsilateral to carotid occlusion, producing both areas of frank scarring and marked atrophy at this timepoint. (A) LM22A-4-treated mouse (B) saline-treated mouse. *, hole made by a needle that was used to mark the non-stroked side prior to sectioning and after sacrifice. 5

Supplementary Figure 2. Horizontal ladder testing on day 1 and rotarod testing on day 2 after surgery correlate significantly with stroke size, and ladder test results are superior. (A) Left front error on the horizontal ladder test. (B) Rotarod performance compared to baseline. n = 14 in hypoxic-ischemic stroke group and 6 shams. P values and R2 values are from linear regression using all mice. 6

Supplementary Figure 3. Angiogenesis is not increased by LM22A-4 treatment. (A) Representative immunostaining for BrdU and PECAM. Scale bar, 20 µm. (B) TrkB pathway stimulation with LM22A-4 did not significantly alter the total number of BrdU+/PECAM+ double-positive cells in affected (dorsolateral) or unaffected (ventral) regions of the striatum, or in penumbral cortex at ten weeks after stroke. n = 10 per group (C) Representative photomicrographs of PECAM immunostaining in penumbral cortex. Scale bar, 200 µm. (D) Quantification of total PECAM immunostaining in penumbral cortex did not reveal significant differences in blood vessel density. n = 7-9 per group; Graphs, means ± SEM. 7

Supplementary Figure 4. LM22A-4 treatment did not affect markers of astrogliosis or immune response at 10 weeks after stroke. (A) Representative photomicrograph of peri-infarct cortex immunostained with GFAP. Graph is percent area covered by GFAP immunostaining. (B) Representative photomicrograph of peri-infarct cortex immunostained with the activated microglial/macrophage marker CD68. Graph is percent area covered by CD68 immunostaining. (C) Representative photomicrograph of peri-infarct cortex immunostained with MHC II. Graph is percent area covered by MHC II immunostaining. Graphs are mean ± SEM. Scale bars, 100 µm. 8

Supplementary Figure 5 - LM22A-4 exerted no effect on neuronal arborization of striatal medium spiny neurons and layer 2/3 or 5/6 motor cortex pyramidal neurons contralateral to stroke. Golgi staining was performed on the contralateral hemisphere of mice sacrificed 72 days after stroke or sham surgery. Motor cortex pyramidal neurons in layers 5/6 and 2/3, and medium spiny neurons in the dorsolateral striatum were traced using Neurolucida software. Five neurons per region per mouse were traced. No significant differences were observed between groups in number of primary dendrites, average dendrite nodes, total dendrite length, or average dendrite ends. n = 6-8 mice per group. Graphs are mean ± SEM. 9

Supplementary Figure 6. LM22A-4 treatment did not increase axonal sprouting after stroke. Mice underwent dMCAO stroke and were then treated with LM22A-4 0.22 mg/kg or saline from days 3-21 after stroke. BDA was injected into barrel cortex contralateral to the stroke on day 14, one week before sacrifice. (A,B) Graphs of quantification of BDA-containing axons in (A) ipsilesional vs. contralesional dorsolateral striatum and (B) penumbral cortex vs. the central commisure of the corpus callosum. Graphs, mean ± SEM.

REFERENCES

1. Yang T, Bernabeu R, Xie Y, Zhang JS, Massa SM, Rempel HC, et al. Leukocyte antigen- related protein tyrosine phosphatase receptor: A small ectodomain isoform functions as a homophilic ligand and promotes neurite outgrowth. J Neurosci. 2003;23:3353-3363 2. Schmid D, Yang T, Ogier M, Adams I, Mirakhur Y, Wang Q, Massa, SM, Longo FM, Katz DM. A TrkB small molecule partial agonist rescues TrkB phosphorylation deficits and improves respiratory function in a mouse model of Rett Syndrome J Neurosci. 2012; 32:1803-1810. 3. Andres RH, Choi R, Pendharkar AV, Gaeta X, Wang N, Nathan JK, et al. The ccr2/ccl2 interaction mediates the transendothelial recruitment of intravascularly delivered neural stem cells to the ischemic brain. Stroke. 2011;42:2923-2931 4. Metz GA, Whishaw IQ. Cortical and subcortical lesions impair skilled walking in the ladder rung walking test: A new task to evaluate fore- and hindlimb stepping, placing, and co-ordination. J Neurosci Methods. 2002;115:169-179 5. Guzman R, De Los Angeles A, Cheshier S, Choi R, Hoang S, Liauw J, et al. Intracarotid injection of fluorescence activated cell-sorted cd49d-positive neural stem cells improves targeted cell delivery and behavior after stroke in a mouse stroke model. Stroke. 2008;39:1300-1306