supplementary materials for2019/12/02  · helsinki institutional review board, and written informed...

25
stm.sciencemag.org/cgi/content/full/11/521/eaaw8283/DC1 Supplementary Materials for Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction Vladimir V. Senatorov Jr., Aaron R. Friedman, Dan Z. Milikovsky, Jonathan Ofer, Rotem Saar-Ashkenazy, Adiel Charbash, Naznin Jahan, Gregory Chin, Eszter Mihaly, Jessica M. Lin, Harrison J. Ramsay, Ariana Moghbel, Marcela K. Preininger, Chelsy R. Eddings, Helen V. Harrison, Rishi Patel, Yishuo Shen, Hana Ghanim, Huanjie. Sheng, Ronel Veksler, Peter H. Sudmant, Albert Becker, Barry Hart, Michael A. Rogawski, Andrew Dillin, Alon Friedman, Daniela Kaufer* *Corresponding author. Email: [email protected] Published 4 December 2019, Sci. Transl. Med. 11, eaaw8283 (2019) DOI: 10.1126/scitranslmed.aaw8283 The PDF file includes: Fig. S1. Age-related BBB function, astrocyte reporter expression, and neurological function in mice. Fig. S2. Age-related BBB function in neurologically intact human subjects. Fig. S3. Characterization of the induced albumin intracerebroventricular infusion rodent model. Fig. S4. Characterization of the astrocytic TGFβR KD mouse line. Fig. S5. Pharmacokinetics of the small-molecule TGFβR inhibitor IPW. Fig. S6. IPW reverses seizure vulnerability in aged mice. Fig. S7. A scheme representation of TGFβ family of ligand and receptors. Table S1. Age and gender of human subjects who received DCE-MRI scans. Table S2. Gender and age of subjects providing postmortem brain tissue. Table S3. Summary of statistical tests used in each experiment. Table S4. PCR primers used to perform genotyping. Table S5. Components of PCR reactions. Table S6. PCR thermocycling conditions. Table S7. Primers for quantitative PCR. References (97102) Other Supplementary Material for this manuscript includes the following: (available at stm.sciencemag.org/cgi/content/full/11/521/eaaw8283/DC1) Data file S1. Tabular data points for experiments with a sample size of n < 20 (provided as a separate Excel file).

Upload: others

Post on 24-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

stm.sciencemag.org/cgi/content/full/11/521/eaaw8283/DC1

Supplementary Materials for

Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling

and chronic yet reversible neural dysfunction

Vladimir V. Senatorov Jr., Aaron R. Friedman, Dan Z. Milikovsky, Jonathan Ofer, Rotem Saar-Ashkenazy, Adiel Charbash, Naznin Jahan, Gregory Chin, Eszter Mihaly, Jessica M. Lin, Harrison J. Ramsay, Ariana Moghbel, Marcela K. Preininger,

Chelsy R. Eddings, Helen V. Harrison, Rishi Patel, Yishuo Shen, Hana Ghanim, Huanjie. Sheng, Ronel Veksler, Peter H. Sudmant, Albert Becker, Barry Hart, Michael A. Rogawski, Andrew Dillin, Alon Friedman, Daniela Kaufer*

*Corresponding author. Email: [email protected]

Published 4 December 2019, Sci. Transl. Med. 11, eaaw8283 (2019)

DOI: 10.1126/scitranslmed.aaw8283

The PDF file includes:

Fig. S1. Age-related BBB function, astrocyte reporter expression, and neurological function in mice. Fig. S2. Age-related BBB function in neurologically intact human subjects. Fig. S3. Characterization of the induced albumin intracerebroventricular infusion rodent model. Fig. S4. Characterization of the astrocytic TGFβR KD mouse line. Fig. S5. Pharmacokinetics of the small-molecule TGFβR inhibitor IPW. Fig. S6. IPW reverses seizure vulnerability in aged mice. Fig. S7. A scheme representation of TGFβ family of ligand and receptors. Table S1. Age and gender of human subjects who received DCE-MRI scans. Table S2. Gender and age of subjects providing postmortem brain tissue. Table S3. Summary of statistical tests used in each experiment. Table S4. PCR primers used to perform genotyping. Table S5. Components of PCR reactions. Table S6. PCR thermocycling conditions. Table S7. Primers for quantitative PCR. References (97–102)

Other Supplementary Material for this manuscript includes the following: (available at stm.sciencemag.org/cgi/content/full/11/521/eaaw8283/DC1)

Data file S1. Tabular data points for experiments with a sample size of n < 20 (provided as a separate Excel file).

Page 2: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Materials and Methods: BBB imaging. The human imaging protocol was approved by the Soroka University Medical Center Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105 subjects with an age range of 21 to 83 years old. MRI scans were performed using a 3T Philips Ingenia scanner, and included: T1-weighted anatomical scan (3D gradient echo, TE/TR = 3.7/8.2 ms, acquisition matrix 432x432, voxel size: 0.5x0.5x1 mm), T2-weighted imaging (TE/TR = 90/3000 ms, voxel size 0.45x0.45x4 mm). For the calculation of pre-contrast longitudinal relaxation times (T10), variable flip angle (VFA) method was used (3D T1w-FFE, TE/TR = 2/10 ms, acquisition matrix: 256x256, voxel size: 0.89x0.89x6 mm, flip angles: 10, 15, 20, 25 and 30°). Dynamic contrast-enhanced (DCE) sequence was then acquired (Axial, 3D T1w-FFE, TE/TR = 2/4 ms, acquisition matrix: 192x187 (reconstructed to 256x256), voxel size: 0.9x0.9x6 mm, flip angle: 20°, Δt = 10 Sec, temporal repetitions: 100, total scan length: 16.7 minutes). An intravenous bolus injection of gadoterate meglumine (Gd-DOTA, Dotarem) was administered using an automatic injector after the first five DCE repetitions. Data preprocessing included image registration and normalization to MNI coordinates (using SPM (http://www.fil.ion.ucl. ac.uk/spm)). BBB permeability was calculated for each brain voxel using in-house MATLAB script, as described(97–99). Briefly, a linear fit is applied to the later part of the concentration curve of each voxel; the slope is then divided by the slope at the superior sagittal sinus, to compensate for physiological (heart rate, blood flow) and technical (contrast agent injection rate) variability. For region specific permeability, disrupted region voxels were divided by total region voxels according to SPM registration. Immunostaining. For human staining, postmortem hippocampus was obtained from young (n=3, mean age = 31.3 ± 5 years) and old patients (n=10, mean age = 70.6 ± 5.6 years). All participants gave informed and written consent and all procedures were conducted in accordance with the Declaration of Helsinki and approved by the University of Bonn ethics committee. Resected hippocampi were fixed in 4% formalin and processed into liquid paraffin. All specimens were sliced at 4 µm with a microtome (Microm), mounted on slides, dried, and deparaffined in descending alcohol concentration. For mouse samples, mice were anesthetized with Euthasol euthanasia solution and transcardially perfused with ice cold heparinized physiological saline (10 units heparin/mL physiological saline) followed by 4% paraformaldehyde (PFA, Fisher Scientific #AC416785000) in 0.1 M phosphate buffered saline (PBS). Brains were removed, post-fixed in 4% PFA for 24 hours at 4° C, and cryoprotected in 30% sucrose in PBS. Brains were then embedded in Tissue-Tek O.C.T. compound (Sakura), frozen, and sliced on a cryostat into 20 µm coronal sections, mounted on slides. Samples were immunostained under the following protocol. Slides were treated for antigen retrieval (for human, 5 min incubation at 100° C in sodium citrate buffer, pH 6.0); for mouse, 15 min incubation at 65 °C in Tris-EDTA buffer (10mM Tris Base, 1 mM EDTA solution, 0.05% Tween 20, pH 9.0), then incubated in blocking solution (5% Normal Donkey Serum in 0.1% Triton X-100/TBS) for 1 hour at room temperature. Samples were then stained with primary antibody at 4° C, followed by fluorescent-conjugated secondary antibody for 1 hour at room temperature, and then incubated with DAPI (900 nM; Sigma-Aldrich) to label nuclei. For human, primary antibodies were rabbit anti-phosphorylated Smad2 (Millipore AB3849-I, 1:500), chicken anti-Albumin (Abcam ab106582, 1:500), and mouse anti-GFAP (Millipore MAB3402, 1:500); for mouse, the same were used except anti-phosphorylated Smad2 (Millipore AB3849) and goat anti-GFAP (Abcam ab53554, 1:1000). Secondary antibodies were anti-rabbit Alexa Fluor 568, anti-chicken Alexa Fluor 647, anti-goat Alexa Fluor 488, anti-mouse Alexa Fluor 488 (1:500, Jackson ImmunoResearch), and anti-goat Alexa Fluor 647 (Abcam ab150131, 1:500). All antibodies dilutions were in blocking solution. All antibodies were validated by manufacturer QC assays (see vendor website for details using provided antibody catalog numbers). For tissue from human patients and aged mice, slide-mounted brain

Page 3: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

sections for treated with TrueBlack Lipofuscin Autofluorescence Quencher (Biotium #23007) before coverslip mounting. Images were acquired at 20X or 40X objective magnification using a Zeiss Axio Observer Research microscope (Carl Zeiss AG) and Metamorph software (version 7.7.7.0), and analyzed using ImageJ software (NIH). For human samples, counts were performed in 10 of randomly selected sampling areas (0.453 mm2) from 1-2 sections per subject, at randomized locations in the hippocampus. For mouse, imaging was performed in at least 3-4 hippocampi per mouse. For each section, this included the area of the dentate gyrus, delimited by the granule cell layer and the end of the CA3 cell layer. Cell counts for each individual marker, and co-labeling, were calculated manually by a observer blinded to condition, and normalized to the area or total number of DAPI-positive cells. For each subject, counts from each sampling area were averaged. Human transcriptome analysis. To investigate TGFβ gene expression patterns in brain hippocampus we evaluated expression data of 123 individuals from the Genotype-Tissue Expression (GTEx) project v7 release (57). RNA sequencing was done by the GTEx consortium to measure the gene expression from samples, reported as transcripts per million (TPM). We obtained data derived from hippocampal tissue samples for expression of the TGFβ isoforms TGFβ1 (ENSG00000105329.5), TGFβ2 (ENSG00000092969.7), and TGFβ3 (ENSG00000119699.3), and performed linear regression and Pearson correlations on the resulting datasets with respect to subject age. Evans Blue assay. 4% Evans Blue (EB) solution in 0.9% sterile saline was administered by i.v. bolus injection through the tail vein at a dose of 2 mL/kg. Thirty minutes after tracer administration, brains were transcardially perfused at a rate of 2 mL/min first with PBS for 5 minutes, then with 4% PFA dissolved in PBS for 10 minutes. The brain was removed, post-fixed, and cryoprotected in 30% sucrose in PBS. Brains were cryosectioned at 20 um and slide mounted using Entellan mounting solution (Millipore). Sections were imaged at 20X objective magnification using an Axio Scan.Z1 slide scanner (Zeiss), an automated scope system in which adjacent images are captured and “stitched” together using the Zen microscope software to assemble a single image spanning the entire brain section. For each section, total number of EB positive cells were quantified within the hilar dentate gyrus region of the hippocampus, and then normalized by region area in order to calculate EB+ cells per mm2 (area traces were drawn and measured using Zen software). 3-5 sections were quantified per animal. RT-qPCR. Total RNA was extracted from 200-250 mg of frozen mouse hippocampal tissue using TRIzol reagent (ThermoFisher Scientific #15596026), and further purified with DNA-free DNA Removal Kit (ThermoFisher Scientific AM1906). First-strand cDNA synthesis was performed from 1 µg isolated RNA template using iScript RT supermix (Bio-Rad #1708841). PCR products were amplified using a CFX96 Real-Time PCR System (Bio-Rad), and threshold cycles were detected using SsoAdvanced Universal SYBR Green Supermix (Bio-Rad #172-5271). Mean threshold cycles were normalized to 18s or GAPDH internal control, and relative gene expression were quantified using the 2-ΔΔCT method. Primer sequences were obtained from the NCI/NIH qPrimerDepot and are listed in Table S7. Western Blot. Mouse or rat hippocampal tissue was homogenized and protein lysates were extracted using RIPA buffer (50 mM Tris-HCl, 150 mM NaCl, 1% NP-40, 0.5% Sodium deoxycholate, 0.1% SDS) including a protease (Calbiochem #539134) and phosphatase inhibitor cocktail (Roche PhoStop Ref: 4906845001). Protein samples were run under reducing conditions. 20 μg of protein lysate was mixed with Laemmli buffer (Bio-Rad #161-0737), containing 5% 2-mercaptoethanol (Sigma M6250), and fractionated by SDS-PAGE using the Mini-PROTEAN Tetra System and pre-cast TGX™ Gels (Bio-Rad #456-1096); Following separation, samples were transferred to a nitrocellulose membrane (0.45 µm, Bio-Rad #1620115). Membranes were blocked for 1 hr at room temperature with 5% non-fat dry milk (Apex #20-

Page 4: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

241) or 5% BSA (Biotium #22013) in TBST (10 mM Tris, 150 mM NaCl, 0.5% Tween 20, pH 8.0), and incubated overnight at 4 °C with primary antibody. Membranes were then washed 3x10 min with TBST and incubated with secondary antibodies for 1 hr at room temperature. Membranes were washed with TBST 3x10min and visualized using chemiluminescence SuperSignal West Dura Extended Substrate (ThermoFisher Scientific #34075), and Bio-Rad Chemidoc system with Bio-Rad Image Lab software (version 4.0.1). Densitometry analysis was done using Image J (NIH). The following primary and secondary antibodies were used: rabbit anti-β-Actin (1:2000, Cell Signaling #4970), rabbit anti-GAPDH (1:2000, Cell Signaling #2118), rabbit anti-TGFβR2 (1:1500, Abcam ab186838), rabbit anti-phosphorylated Smad2 (1:1000, Millipore AB3849-I), rabbit anti-phosphorylated Smad2 (1:1000, Millipore AB3849), anti-rabbit TGFβ1 (1:500, ab92486), anti-rabbit HRP (1:2000, Cell Signaling #4970). Osmotic pump implants. For mice, pumps were implanted as previously described (100). Briefly, surgery was performed on adult male mice under isofluorane anesthesia (2%). Using a stereotaxic frame, a hole was drilled through the skull at 0.5 mm posterior, 1 mm lateral to bregma, and a cannula was placed into the right lateral cerebral ventricle, fixed with surgical glue. Cannulas (Brain infusion kit BIK 3, #0008851, Alzet) were attached to micro-osmotic pumps (Model 2001, ALZET) filled with 200 μL of either 0.4 mM bovine serum albumin (Alb; Sigma-Aldrich) solution or artificial cerebrospinal fluid (aCSF) as previously described (101), and implanted subcutaneously in the right flank. In a subset of animals, 10% of the Alb was replaced with Alexa Fluor 647 conjugated BSA (2.68 g/L; ThermoFisher Scientific A34785). Pumps infused at a rate of 1.0 µL/hr for the duration described in each experiment. For rats, 10 week-old male Wistar rats were used, and surgeries were performed the same way, using the following coordinates: -1 mm posterior and 1.5 mm lateral to bregma. For rats, albumin was used at a concentration of 0.2 mM and infused for 7 days at a rate of 10 μL/hr via larger micro-osmotic pumps (Model 2ML1, Alzet). ECoG. ECoG was recorded as previously reported (50) from 9- to 12-wk-old Wistar male rats implanted with osmotic pumps, and from young (3 months, n = 5) and old (18-22 months, n = 20) mice. In brief, under stereotaxic surgery and 2% isoflurane anesthesia, two screw electrodes were implanted in each hemisphere (rat coordinates: 4.8 mm posterior, 2.7 mm anterior, and +/- 2.2 mm lateral for all screws; mouse coordinates: 0.5 and 3.5 mm posterior and +/- 1 mm lateral for all screws, all relative to bregma). A wireless transmitter (Data Science International, Saint Paul, MN, US) was placed in a dorsal subcutaneous pocket, and leads connected to the screws. Connections were isolated and fixed by bone cement such that one ECoG channel was associated with each hemisphere. For rat surgeries, a cannula (-1 mm posterior to bregma, 1.5 lateral, 4 mm depth) and osmotic pump (infusing at 2.4 μl/h) were also implanted (28). Animals were treated with post-operative buprenorphine (0.1 mg/Kg) and allowed to recover for at least 72 hrs prior to recording. Continuous, bichannel ECoG (sampling rate, 500Hz) was recorded wirelessly from freely roaming animals in the home cage for the duration of experiments described. Rats were recorded during the first and the fourth week following surgery. Mice were recorded for 15 days, starting 1 week after surgery, and administered IPW (20 mg/kg) on days 6-10 of recording. To detect PSWE, ECoG signals were buffered into 2 sec long epochs with an overlap of 1 sec. Fast Fourier Transform (FFT) was applied and the frequency of median power was extracted for each epoch. Thus, an event was considered as a PSWE if its frequency of median power was less than 5 Hz for 10 consecutive seconds or more. FFT was also applied for the entire recording period to analyze relative power across the frequency spectrum of 1-20 Hz. ECoG signal was filtered using a 1 Hz high pass filter and 100 Hz low pass filter. For iAlb rats, infusion cannulas were placed unilaterally and thus ECoG signal from ipsilateral and contralateral hemispheres were analyzed separately; for young and old mice, no differences were observed between hemispheres and so data from right hemisphere only was used in plots and analysis.

Page 5: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Animal care and transgenic mice. All animal procedures were approved by the institutional animal care committees. Animals were housed with a 12:12 light:dark cycle with food and water available ad libitum. Aldh1L1-eGFP mice were bred from STOCK Tg(Aldh1l1-EGFP)OFC789Gsat/Mmucd (identification number 011015-UCD), was obtained from the Mutant Mouse Resource and Research Center (MMRRC) at University of California at Davis, an NIH-funded strain repository, and was donated to the MMRRC by Nathaniel Heintz, Ph.D., The Rockefeller University, GENSAT. These FVB/N mice were crossed to a C57BL/6 genetic background. The resulting strain exhibits constitutive astrocytic expression of eGFP protein under the astrocytic promoter Aldh1L1. Triple transgenic aTGFβR/KD mice were bred from strains purchased from the Jackson Laboratory to generate mice that express CreERT under the astrocytic promoter glial high affinity glutamate transporter (GLAST), with a floxed exon 4 of TGF-βR2 (tgfbr2fl), and a transgenic LacZ reporter gene inhibited by a floxed neomycin cassette. Tamoxifen induction thus induces activation of astrocytic CreERT resulting in a null TGFβR2 allele (tgfbr2null) and LacZ expression (R26R-/-). The parental strain STOCK Tg(Slc1a3-cre/ERT)1Nat/J mice were outcrossed with B6;129-Tgfbr2tm1Karl/J and B6.129S4-Gt(ROSA)26Sortm1Sor/J mice to produce males, while B6;129-Tgfbr2tm1Karl/J and B6.129S4-Gt(ROSA)26Sortm1Sor/J mice were outcrossed to produce females. The resulting GLAST-CreERT; tgfbr2fl/+ males were bred with tgfbr2fl/+; R26R-/- and tgfbr2fl/fl; R26R-/- females to produce triple transgenic offspring. Subsequent generations were incrossed to produce experimental triple transgenic mice of genotypes GLAST-CreERT; tgfbr2fl/fl; R26R-/-, GLAST-CreERT; tgfbr2fl/+; R26R-/-, GLAST-CreERT; tgfbr2fl/fl; R26R-/+, and GLAST-CreERT; tgfbr2fl/+; R26R-/-. All mice were genotyped via PCR analysis of tissue biopsy samples (Table S4-S6). The inducible Cre/lox system was activated by 5 days of tamoxifen injection (Sigma-Aldrich, 160 mg/kg dissolved in corn oil, i.p.). Control GLAST-CreERT; tgfbr2fl/+ heterozygotes received the same dosage of tamoxifen and control GLAST-CreERT; tgfbr2fl/fl mice received i.p. injection of corn oil vehicle at equivalent volumes. Mice were weighed daily to ensure accurate dosage. Seizure induction. Seizures were induced by a single injection of pentylenetetrazole (PTZ; Sigma, P6500, 85 mg/kg s.c.). Video recordings were taken for 30 minutes following induction, which were then scored by a blind observer, using a modified Racine scale to quantify progression of seizure severity, as follows: 0 – No Seizures; 1 – Immobility; 2 – Straub’s tail; 3 – Forelimb Clonus; 4 – Generalized Clonus; 5 – Bouncing Seizure; 6 - Status Epilepticus. Latency to first appearance of each Racine stage was quantified, as was latency to mortality (when occurring prior to the 30 min endpoint). Behavior assessments. Spatial working memory was tested by spontaneous alternation in a T-maze constructed of black plastic, with stem (50 x 16 cm) and two arms (50 x 10 cm). A vertical divider was placed at the midline of the stem exit, thus creating two entryways leading to either the right or left arms. Naïve mice were placed at the beginning of the stem and allowed to freely roam until opting to enter either the right or left arm. Upon crossing the threshold of the chosen arm, a door was lowered, and the mouse was contained to the chosen arm for 30 sec. Then, the mouse was returned to the stem and the door raised, allowing the next choice trial to commence immediately. After 10 sequential trials, the mouse was returned to the home cage. “Correct” alternation choices were scored when the mouse chose the arm opposite of that chosen in the previous trial, and percent correct was calculated as (total correct choices) / (total number of completed trials). In the event that a mouse did not leave the stem within 60 sec, it was removed from the apparatus for 30 sec, and then reset in the stem to start a new trial. These “incomplete” trials were not counted in scoring. Spatial memory in young mice was assessed

Page 6: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

in the Morris Water Maze (MWM) task (102). Because of excess colony availability, heterozygous control mice from the transgenic colony (TGFβR2 (fl/+) genotype), which retain normal TGFβ signaling, were used for the MWM study cohort. For each trial, mice were placed at randomized starting locations in a MWM pool filled with opaque water (colored with non-toxic white acrylic paint), with visual cues placed on the pool walls. Mice were allowed to swim freely until locating a hidden platform under the surface of the water (or guided to the platform after 40 seconds of failed searching), and then left on the platform for 10 seconds prior to starting a new trial. Spatial learning was quantified by measuring latency to reach the platform, averaged from four trials per day, with training over 9 consecutive days of learning. Object memory was tested in the novel object task, using a 3-day protocol consisting of a 10 min trial on each day, recorded by overhead video. On day 1 mice were habituated to a square testing chamber, constructed of white plastic (50 x 50 cm), with two unfamiliar objects placed inside. On day 2, the previous objects were removed and 3 new objects were placed in a “L” configuration, equidistant from each other and the walls. On day 3, one of the objects was removed and replaced by a new object, thus leaving 2 familiar objects and 1 novel object. A blind observer quantified duration of time spent investigating each object (scoring criteria were mouse nose oriented towards object at a distance of 1 cm or less), and percentage of time at the novel object was quantified as (duration investigating novel object) / (total duration investigating all three objects). The set objects were chosen from common items such as lab bottles, pipette boxes, cups (placed open-side down), etc., that were of similar dimensions but varied in shape, color, and material. The sequence and position of objects used across trials was identical for all mice. IPW pharmacokinetics. Brain concentration measurement of IPW-5371 was performed by Jubilant Biosys. The experiment was approved by the Jubilant Biosys Institutional Animal Ethics Committee, Bangalore, India (IAEC/JDC/2015/72) and were in accordance with the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), Ministry of Social Justice and Environment, Government of India. Twelve Balb/C mice (age 6-7 weeks) were procured from Bioneeds, Bangalore, India. Animals were housed in Jubilant Biosys animal care facility in a temperature and humidity controlled room with a 12:12 h light:dark cycles, had free access to food (Provimin) and water for one week before experimental use. Following ~4 h fasting (during fasting period animals had free access to water) mice received IPW-5371 orally at a dose of 20 mg/kg (dissolved in 0.5% methylcellulose and saline). Mice were euthanized under isofluorane and the brain was removed and weighed. Brain tissue homogenates were prepared with 10% tetrahydrofuran in acetonitrile (tissue was homogenated with 4 mL/g of 10% tetradhydrofuran in acetonitrile containing IS (100 ng/mL)). Subsequently, 250 µL of brain homogenate was centrifuged at 14,000 rpm for 5 min at 10 °C. An aliquot of 10 µL was injected onto an API 4000 LC-MS/MS system for analysis.

Page 7: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105
Page 8: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S1. Age-related BBB function, astrocyte reporter expression, and neurological function in mice. (A) Percentage of cells (DAPI+) in the mouse hippocampus that colocalize with albumin at given timepoints across the mouse lifespan; albumin extravasation and localization to brain cells increases significantly with age (ANOVA, p < 0.0001, with Bonferroni posttest). (B-C) BBB breakdown in aging mice was confirmed by the Evans blue (EB) assay. 30 min after i.v. EB injection, the EB tracer was mostly contained within blood vessels in young mice (2-4 month old), while middle aged (12 month old) and old (21-24 month old) mice showed significantly elevated extravascular EB tracer localized to cells (ANOVA, n = 5, p < 0.0002, with Dunnett’s posttest). Representative images from mouse hilar hippocampal dentate gyrus show EB (purple) localized to blood vessels (BV, yellow arrows) and extravascular EB (EB, white arrows) localized to cells. Scale bar = 20 μM. (D) Percent of total cells expressing fluorescent reporter (Rep) in the hippocampus of Rep-Aldh1L1 mice. The number of labeled astrocytes was quantified across the lifespan. (E) Co-staining for cell-specific markers was used to estimate the percent of albumin taken up by different cell types in the aged mouse hippocampus, as a percent of all albumin positive cells, marked by astrocytes (Reporter gene, REP), microglia (Iba1), oligodendrocytes (CAII), and neurons (NeuN). (F) A modified Racine scale, measuring seizure severity on a scale of 1-6, was used to score seizure vulnerability following injection of PTZ. (onset of first seizure at each stage of severity; 2-way ANOVA, main effect of seizure progression p < 0.0001; main effect of age p = 0.005, n = 13 (3 mo), 10 (12 mo), 8 (18 mo and 24 mo)). For each group, linear regression was fit to the Racine progression, and the slope of the best fit line was used as a measure of seizure vulnerability in Fig. 1F. (G) Spectral analysis of ECoG in aged compared to young mice. This slow-wave activity was examined in greater detail by quantifying discrete PSWEs (see Fig. 1H-I). (H) Circadian plot showing relative occurrence of PSWEs throughout the day. Young mice show a circadian oscillation, with the highest percentage of all PSWEs occurring after onset of the light cycle, whereas hPSWE mice show PSWEs consistently throughout both day and night. (I) GMM clustering was performed to segregate the old mice into lPSWE (asymptomatic) and hPSWE (symptomatic) subgroups. (J) Akaike information criterion (AIC) was used to compare the goodness-of-fit of GMM clustering models with 1, 2, or 3 clusters. Lower AIC values represent better goodness-of-fit, indicating the old mice are best described by the two cluster model (low and high subgroups) rather than as one continuous group. (K) PSWEs quantified in young, old hPSWE and lPSWE groups (ANOVA).

Page 9: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S2. Age-related BBB function in neurologically intact human subjects. (A) Linear regression was used to model the relationship between age and BBB permeability. Percent of total brain volume with permeability values above the 95th percentile of the averaged young healthy subjects (n = 113, p=0.0315). (B) Descriptive statistics estimating the percent of individuals affected by BBB-D (defined as suprathreshold permeability in more than 5% all brain voxels) across age groups.

Page 10: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S3. Characterization of the induced albumin intracerebroventricular infusion rodent model. (A, top) Diagrams showing mouse icv pump implants, with cannulas placed to infuse albumin into the brain ventricle; (A, bottom) representative immunofluorescent image of ipsilateral hemisphere, showing hippocampus (coronal slice). The infused albumin (fluorescent-tagged, in red) diffuses into the ventricle

Page 11: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

and accumulates in the dentate gyrus (arrowhead indicates the granule cell layer). Scale bar = 700 μm. (B) Representative images from the dentate gyrus show albumin uptake in astrocytes at 24 hours, 48 hours, and 7 days after pump implant (with aCSF control shown at 48 hours; scale bar = 50 μm). (C) Progression through Racine scale Following PTZ injection in iAlb and aCSF control mice (2-way ANOVA, main effect of seizure progression p < 0.0001; main effect of treatment p = 0.0001, n = 4). Best fit lines through the Racine progressions were used to measure seizure vulnerability in Fig. 2B. (D-E) Representative ECoG traces from ipsilateral and contralateral hemisphere, showing discrete PSWEs within a 10 second window (marked with *). (F-G) Spectrum analysis of ECoG activity in the ipsilateral and contralateral hemispheres receiving iAlb infusion, Frequency ranges (inset graph) are δ = 1-5 Hz, θ = 5-8 Hz, α = 8-12 Hz, and β = 12-20 Hz.

Page 12: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105
Page 13: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S4. Characterization of the astrocytic TGFβR KD mouse line. (A) Representative immunofluorescent image from mouse hippocampus, showing reporter expression (Rep-βGal, red) in astrocytes (GFAP, green) following induction of the Cre recombinase system to knock down (KD) aTGFβR. Scale Bar = 30 μm. (B) Reporter expression in hippocampal astrocytes following induction of KD, quantified throughout all hippocampal subregions (CA: cornu ammonis; ML: molecular layer; SGZ: subgranular zone). (C) Representative images of reporter expression with cell linage markers. No significant reporter expression was observed in mature neurons (NeuN), indicating that hippocampal neural stem cells expressing GLAST did not generate an appreciable lineage of recombinant TGFβR KD neurons. Representative image of reporter expression in mouse hippocampus stained with NeuN to assess neural expression, scale bar = 50 μm. (D) Western blot with densitometry of TGFβR2 in the hippocampus following tamoxifen induction in homozygous KD mice (aTGFβR2 fl/fl) compared to controls (aTGFβR2 +/+) (t-test, p=0.045, n=3). (E) WB from 17-24 month aged mouse hippocampus quantifying pSmad2 amount following KD (t-test, p < 0.0001, n = 9 Tgfbr2+/+, 7 Tgfbr2fl/fl). (F) Quantitative real-time PCR from aged mouse hippocampus quantfying expression of TGFβR2 following KD (t-test, p = 0.036, n = 10 Tgfbr2+/+, 8 Tgfbr2fl/fl). (G-H) Progression through the Racine scale following PTZ test in early and late aged mice (2-way ANOVA; for early aged group, main effect of seizure progression p < 0.0001; main effect of genotype p = 0.0053; interaction p = 0.0214, with Bonferroni posttest, n = 5 Tgfbr2+/+, 6 Tgfbr2fl/+ and Tgfbr2fl/fl; for late aged group, main effect of seizure progression p < 0.0001; main effect of genotype p < 0.0001; interaction p = 0.026). Best fit lines of Racine progression were used to measure seizure vulnerability in Fig. 3C and D.

Page 14: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S5. Pharmacokinetics of the small-molecule TGFβR inhibitor IPW. (A) PK measures of IPW concentration in brain lysates from n = 3 mice at timepoints spanning 24 hours after a single dose (30 mg/kg, po). At 24 hours, brain IPW concentration remains above the IC50 of 75 nM. (B) PK measures of IPW concentration in blood from n = 3 mice at timepoints spanning 24 hours, and an extended 48 hour timepoint. After 24 hours, IPW concentration falls below the IC50 of 75 nM (po, oral dosing; iv, intravenous dosing). (C) Progression through the Racine seizure scale following PTZ induction in iAlb mice injected with IPW or vehicle control (2-way ANOVA, main effect of seizure progression p < 0.0001; main effect of treatment p = 0.0001, n = 4 per group). Best fit lines of Racine progression were used to measure seizure vulnerability in Fig. 5C.

Page 15: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S6. IPW reverses seizure vulnerability in aged mice. (A) Progression through the Racine scale following PTZ induction in aged mice treated with IPW or vehicle control for 7 days (2-way ANOVA, main effect of seizure progression p < 0.0001; main effect of drug treatment p < 0.0001; interaction p = 0.0001, with Bonferroni posttest, n = 9). Best fit lines of Racine progression were used to measure seizure vulnerability in Fig. 6D. (B) Number of PSWES quantified in old hPSWE in a longitudinal follow-up of 5 dyas of baseline, 5 days injection of vehicle control and 5 days of washout. (no effect of injection). (C) Average number of PSWEs for all groups during the final two days of each treatment period (Baseline prior to injection, treatment with daily injection of IPW or vehicle control, and washout with cessation of injections).

Se

izu

r e la

t en

cy (

min

) ****

Seizure grade

C

Severity

Old h-SWE + IPW Old l-SWE + IPW

Young + IPW Old h-SWE + vehicle

Old h-SWE + vehicle

Baseline Treatment Washout

A BA

ve

rage

nu

mb

er

of

SW

Es **

****

Treatment

Baseline

Washout

1500

1000

500

0

Treatment

Baseline

Washout

Treatment

Baseline

Washout

Treatment

Baseline

Washout

1500

1000

500

0

Nu

mb

er

of

SW

Es

1 2 3 4 5 6 7 1098 12 13 14 1511

Page 16: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Fig. S7. A scheme representation of TGFβ family of ligand and receptors. The three major members of

the TGFβ ligand family (TGFβ 1, 2, or 3) bind and activate TGFβ Receptors (TGFβ R), a heteromeric

complex consistent of two units – TGFβR1 and TGFβR2. Similarly, albumin binds and activates the

complex TGFβR1/TGFβR2. Activation leads to phosphorylation of the Smad protein (in astrocytes

activation of ALK5 leads to phosphorylation of Smad2), which in turn induce transcriptional modulation,

and an increase in TGFβ1 transcription and release, in a positive feedback mechanism. Loss of function

was induced here in a transgenic model by knockdown of TGFβR subunit II in astrocytes, and

pharmacologically, using the small molecule TGFβR1 kinase inhibitor, IPW. B. Schematic summarizing

the mechanistic model underlying BBB-related neural pathology. In old age, serum proteins including

albumin cross the dysfunctional BBB and activate astrocytic TGFβ receptor (TGFβR) signaling. Outputs of

the astrocytic TGFβ signaling cascade include positive feedback and induction of symptomatic pathology

including aberrant neural activity, hyperexcitability, and cognitive impairment.

Page 17: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Table S1. Age and gender of human subjects who received DCE-MRI scans.

Age Gender MRI

abnormalities Diagnosis*

18.91 M None None

22.17 F None None

24 M None None

25 F None None

25 M None None

25 M None None

25.71 M None None

26 M None None

26 M None None

26 M None None

26 M None None

26.12 F None None

26.6 F None None

26.67 F None None

27 M None None

27.24 F None None

28 M None None

28 M None None

28 M None None

28 M None None

28.59 M None None

29.46 M None None

30 M None None

30 F None None

30 M None None

30 M None None

30 M None None

30.37 F None None

30.53 F None None

30.56 M None None

30.69 M None None

31 M None None

32 M None None

32 M None None

32.06 M None None

32.55 F None None

33.24 F None None

33.72 M None None

39 M None None

Page 18: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

40.91 M None None

42 M None None

42.46 M None None

43 F None None

44 F None None

44.33 M None None

45.63 F None None

47.62 F None None

48 F None None

49 F None None

49 F None None

51 M None None

51.58 F None None

52.68 F None None

54 F None None

54.16 F None None

55 F None None

56 M None None

57 F None None

57.22 F None None

59.63 F None None

60 F None None

62 F None None

64 F None None

64.22 F None None

64.85 F None None

67 M None None

67 M None None

67.18 M None None

68.3 M None None

68.97 F None None

69.41 F None None

70 F None None

70.18 F None None

70.81 M None None

72 F None None

73.02 M None None

76.4 F None None

76.89 F None None

80 M None None

24 M None None

25 M None None

26 M None None

26.36 M None None

Page 19: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

27 M None None

28 M None None

28 M None None

28.39 M None None

29 M None None

31 M None None

33 M None None

35.13 F None None

37 F None None

41 M None None

43 F None None

48 F None None

49.16 F None None

50 F None None

52.02 F None None

55 M None None

55 F None None

56 M None None

58 F None None

59 F None None

59 M None None

59.4 M None None

60 M None None

65 M None None

68 F None None

70 M None None

75 F None None

80 M None None

82 M None None

85 M None None

85.17 M None None

*Patients were screened for confounding neurological conditions, including previous diagnosis of stroke, head injury, and cognitive impairment.

Page 20: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Table S2. Gender and age of subjects providing postmortem brain tissue.

Gender Age Diagnosis*

m 26 None

m 32 None

f 36 None

m 61 None

f 63 None

m 68 None

f 69 None

m 70 None

m 71 None

m 74 None

f 75 None

m 77 None

m 78 None

*Subjects were screened for any confounding neurological condition, including stroke, head injury, and

dementia/neurodegenerative disease.

Page 21: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Table S3. Summary of statistical tests used in each experiment.

Experiment Figure Sample size (n) Statistical test p value

Albumin + pSmad counts (aging timecourse)

1A-C 3 mo old = 4 12 mo old = 5 18 mo old = 3 21 mo old = 6

ANOVA w/ Bonferroni posttest

<0.0001

pSmad2 & TGFβ WB (young vs old)

1E 5 T-test w/ Welch's correction

pSmad2 = 0.018 TGFβ = 0.018

PTZ seizure resilience (aging timecourse)

1F-G 3 mo old = 13 12 mo old = 10 18 mo old = 8 21 mo old = 8

ANOVA w/ Bonferroni posttest

0.001

ECoG recordings of PSWE (young vs old)

1H-I young = 5 old = 18

Mann-Whitney test

0.019

DCE-MRI of BBB permeabillity in aging human subjects

2A-B young = 59 old = 29

T-test 0.001

Albumin + pSmad counts (aging human)

2C-D young = 3 old = 10

T-test 0.032

TGFβ expression (aging human transcriptome)

2E 123 Pearson correlation

TGFβ1 p = 0.031 TGFβ3 p = 0.042

PTZ asssay seizure resilience and survival (iAlb)

3B-C 4 T-test Resilience p = 0.0497; survival p = 0.004

ECoG PSWE counts (iAlb)

3D aCSF = 5 iAlb = 8

Mann-Whitney test

p = 0.007

MWM learning (iAlb) 3E aCSF = 9 iAlb = 10

Repeated measure ANOVA w/ Bonferroni posttest

Group main effect p = 0.009

PTZ asssay seizure resilience and survival (aTGFβR KD early aging)

4C Tgfbr2+/+ = 5 Tgfbr2fl/+ = 6 Tgfbr2fl/fl = 6

ANOVA w/ Bonferroni posttest

Resilience p = 0.001; survival p = 0.022

PTZ asssay seizure resilience and survival (aTGFβR KD late aging)

4D Tgfbr2fl/+ = 5 Tgfbr2fl/fl = 9

T-test w/ Welch's correction

Resilience p = 0.002; survival p = 0.004

Page 22: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

T-maze (aTGFβR KD early aging)

4E Tgfbr2fl/+ = 21 Tgfbr2fl/fl = 20

Mann-Whitney test

0.028

T-maze (aTGFβR KD late aging)

4F Tgfbr2fl/+ = 12 Tgfbr2fl/fl = 11

Mann-Whitney test

0.035

T maze correlation with pSmad2 amount (aTGFβR KD)

4G 14 Pearson correlation

0.024

iAlb pSmad2 counts 5A-B 6 ANOVA w/ Neuman-Keuls posttest

pSmad2 p = 0.027; pSmad2+rep p = 0.005

PTZ asssay seizure resilience and survival (iAlb)

5C-D aCSF = 6 iAlb + veh = 6 iAlb + IPW = 7

ANOVA w/ Bonferroni posttest

Resilience p = 0.004; survival p = 0.003

pSmad2 counts (aged IPW treatment)

6A old + veh = 5 old + IPW = 6

T-test pSmad2 p = 0.037; pSmad2+Alb+rep p = 0.014

pSmad2 WB (aged IPW treatment)

6B 4 ANOVA w/ Bonferroni posttest

0.0001

TGFβ1 WB (aged IPW

treatment)

6C 4 ANOVA w/ Bonferroni posttest

<0.0001

PTZ asssay seizure resilience and survival (aged IPW treatment)

6D 9 T-test Resilience p < 0.0001; Survival p < 0.0001

ECoG PSWE counts (longitudinal IPW treatment)

6E hPSWE = 6 lPSWE = 6 young = 5

Dunn's multiple comparison

0.0098

T maze (aged IPW treatment)

6F old + veh = 12 old + IPW = 14

T-test 0.028

Novel object task (aged IPW treatment)

6G old + veh = 10 old + IPW = 8

T-test 0.017

Page 23: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Table S4. PCR primers used to perform genotyping.

Primer Sequence Amplicon Size (bp)

Tg(Slc1a3-cre/ERT)1Nat fwd 5' ACA ATC TGG CCT GCT ACC AAA GC 3' Transgene: ~600

Tg(Slc1a3-cre/ERT)1Nat rev 5' CCA GTG AAA CAG CAT TGC TGT C 3' Internal positive control: 200

Tg(Slc1a3-cre/ERT)1Nat IPC 1 5' CAA ATG TTG CTT GTC TGG TG 3'

Tg(Slc1a3-cre/ERT)1Nat IPC 2 5' GTC AGT CGA GTG CAC AGT TT 3'

Tgfbr2tm1Karl fwd 5' TAT GGA CTG GCT GCT TTT GTA TTC 3' Mutant: 575

Tgfbr2tm1Karl rev 5' TGG GGA TAG AGG TAG AAA GAC ATA 3' Heterozygote: 415 and 575

Wild type: 415

Gt(ROSA)26Sortm1Sor common 5' GCG AAG AGT TTG TCC TCA ACC 3' Mutant: 340

Gt(ROSA)26Sortm1Sor mutant rev 5' AAA GTC GCT CTG AGT TGT TAT 3' Heterozygote: 340 and ~650

Gt(ROSA)26Sortm1Sor wild type rev 5' GGA GCG GGA GAA ATG GAT ATG 3' Wild Type: ~650

eGFP fwd 5' CCT ACG GCG TGC AGT GCT TCA GC 3' eGFP: ~300

eGFP rev 5' CGG CGA GCT GCA CGC TGC GTC CTC 3'

Page 24: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Table S5. Components of PCR reactions.

Allele Reaction Component Volume per Reaction (μL)

Tg(Slc1a3-cre/ERT)1Nat

Forward Primer 4

Reverse Primer 4

IPC 1 Primer 2

IPC 2 Primer 2

Apex 2X Taq RED

Master Mix 12.5

DNA Template 0.5

Total 25

Tgfbr2tm1Karl

Forward Primer 2.5

Reverse Primer 2.5

Apex 2X Taq RED

Master Mix 12.5

Water 7

DNA Template 0.5

Total 25

Gt(ROSA)26Sortm1Sor

Common Primer 2.5

Mutant Reverse Primer 2.5

Wild Type Reverse

Primer 2.5

Apex 2X Taq RED

Master Mix 12.5

Water 4

DNA Template 0.5

Total 25

eGFP

Forward Primer 2.5

Reverse Primer 2.5

Apex 2X Taq RED

Master Mix 12.5

Water 7

DNA Template 0.5

Total 25

Page 25: Supplementary Materials for2019/12/02  · Helsinki institutional review board, and written informed consent was given by all participants. BBB status was assessed by DCE-MRI in n=105

Table S6. PCR thermocycling conditions.

Allele Cycling Step # Temp (°C) Time Note

Tg(Slc1a3-cre/ERT)1Nat

1 94 3:00 -

2 94 0:30 -

3 60 0:30 -

4 72 0:30 -

5 - - Go to 2, 35X

6 72 2:00 -

7 12 0:00 infinite hold

Tgfbr2tm1Karl

1 94 3:00 -

2 94 0:30 -

3 62 0:30 -

4 72 0:30 -

5 - - Go to 2, 35X

6 72 2:00 -

7 10 0:00 infinite hold

Gt(ROSA)26Sortm1Sor

1 98 3:00 -

2 98 0:30 -

3 65 1:00 -

4 72 1:00 -

5 - - Go to 2, 35X

6 72 2:00

7 10 0:00 infinite hold

eGFP

1 94 3:00 -

2 94 0:30 -

3 60 0:45 -

4 72 0:45 -

5 - - Go to 2, 35X

6 72 10:00 -

7 11 0:00 infinite hold

Table S7. Primers for quantitative PCR.

Tgfbr2-F CTGGCCATGACATCACTGTT

Tgfbr2-R GTCGGATGTGGAAATGGAAG