Neural crest-derived brain pericytes (iBPC)
Human induced pluripotent stem cells (hiPSC) were differentiated to neural crest (NC)-derived brain pericytes (iBPC) using previously published protocols (43, 44). Briefly, Gibco episomal hiPSC line (Gibco, #A13700, Thermo Fisher Scientific, Leusden, The Netherlands) was cultured in mTeSR Plus medium (STEMCELL Technologies, Vancouver, Canada) on vitronectin-coated plates (Invitrogen, Thermo Fisher Scientific, Leusden, The Netherlands). To start the differentiation, hiPSCs were passaged as single cells and seeded at a density of 2 x 105 cells/cm2 onto Matrigel-coated plates and cultured in NC induction medium, consisting of DMEM/F12 GlutaMAX™ (Gibco, Thermo Fisher Scientific, Leusden, The Netherlands), 1× B27 (Gibco, Thermo Fisher Scientific, Leusden, The Netherlands), 0.5% BSA and 3 µM CHIR 99021 (Tocris, Bristol, United Kingdom) for 5 days. The resulting NC cells were passaged as single cells and seeded at a density of 2.5 x 104 cells/cm2 onto 0.1% gelatin-coated plates and cultured in pericyte medium (ScienCell, Carlsbad, CA, USA) for 5 days for BPC specification. Immunocytochemistry and mRNA evaluation of PDGFRβ, NG2, CD13, FOXF2, FOXC1, CD146, vitronectin confirmed their pericyte identity. iBPC were used for sprouting experiments between passages 2 and 4.
Lentiviral short hairpin RNA for LXRα, LXRβ and HIF-1α knockdown
Selective gene knockdown was obtained by using a vector-based short hairpin RNA (shRNA) technique as previously described (45). Recombinant lentiviruses were produced by co-transfecting sub-confluent HEK293T cells with the specific expression plasmids and packaging plasmids (pMDLg/pRRE, pRSV-Rev and pMD2G) using calcium phosphate as a transfection reagent. HEK293T cells were cultured in Dulbecco’s modified eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS) and 1% penicillin/streptomycin. Cells were cultured at 37°C in 5% CO2. Infectious lentiviral particles were collected 48 hours after transfection and stored at − 80°C upon further use. The knockdown (KD) efficiency of all 5 constructs for each gene was tested, and the most effective constructs were used in subsequent experiments. For LXRα (NR1H3), TRC22237 was selected, with encoding sequence 5’-GTGCAGGAGATAGTTGACTTT-3’ that targets nucleotides 1043–1063 of the NM_005693.3 RefSeq. For LXRβ (NR1H2) the most effective construct was TRC275326, encoding the sequence 5’-GAAGGCATCCACTATCGAGAT-3’ that targets nucleotides 1193–1213 of the NM_007121.5 RefSeq. For HIF-1A (HIF1A) the most effective construct was TRCN0000010819, encoding the sequence 5’-TGCTCTTTGTGGTTGGATCTA-3’. Subsequently, lentiviruses expressing LXRα, LXRβ or HIF-1α specific shRNA were used to transduce hCMEC/D3 cells. Control cells were generated by transduction with lentivirus expressing non-targeting shRNA (SHC002, Sigma-Aldrich, St Louis, MO). Twenty-four hours after infection of hCMEC/D3 cells with the shRNA-expressing lentiviruses, stable cell lines were selected by puromycin treatment (2 µg/ml, Sigma-Aldrich, Diegem, Belgium). The knockdown efficiency was determined by quantitative real-time PCR (qRT-PCR) and Western blot. The primer sequence is listed in Supplementary Table. 1. At the start of the experiment, transduced hCMEC/D3s were treated with the LXRs agonist GW3965 (1µM, Sigma-Aldrich, Diegem, Belgium), LXRs antagonist GSK2033 (1µM, R&D systems, Minneapolis, MN, USA), retinoic acid (5µM, Sigma-Aldrich, Diegem, Belgium), RXRα antagonist PA452 (1µM, MCE, New Jersey, USA) or dimethyl sulfoxide (DMSO) as vehicle control for 48 or 72 hours in EGM-2 media (Lonza, Basel, Switzerland).
RNA isolation and qRT-PCR
Recombinant hCMEC/D3 cells (1x106 cells/ml) transduced with either LXRα shRNA, LXRβ shRNA, or non-targeting shRNA were seeded in 24-well plates in culture medium. RNA was isolated using the TRIzol® method (Life Technologies, Bleiswijk, The Netherlands) and cDNA was synthesized using the Reverse Transcription System kit (Promega, Leiden, The Netherlands). Sequences of primers used are listed in Supplementary Table. 1. Quantitative Reverse Transcriptase PCR (qRT-PCR) was carried out using SYBR green master mix (Applied Biosystems, Waltham, MA, USA) and a Step One Plus detection system (Applied Biosystems). Quantification of gene expression was accomplished using the comparative cycle threshold method. Expression levels were normalized to Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) or Ribosomal Protein Lateral Stalk Subunit P0 (RPLP0) expression.
RNA sequencing-based transcriptional profiling and analysis
The hCMEC/D3 cells were cultured and treated as described above. Total RNA was extracted using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and converted into strand-specific cDNA libraries using the TruSeq Stranded mRNA sample preparation kit (Illumina, San Diego, CA, USA) according to the manufacturer's instructions. Briefly, polyadenylated RNA was enriched using oligo-dT beads and subsequently fragmented, random primed and reverse transcribed using SuperScript II Reverse Transcriptase (Invitrogen, Carlsbad, CA, USA). Second strand synthesis was performed using Polymerase I and RNaseH with replacement of dTTP for dUTP. The generated cDNA fragments were 3' end adenylated, ligated to Illumina paired-end sequencing adapters, and subsequently amplified by 12 cycles of PCR. The libraries were analysed on a 2100 Bioanalyzer using a 7500 chip (Agilent, Santa Clara, CA, USA) and subsequently sequenced with 65 base single reads on a HiSeq2500 using V4 chemistry (Illumina, San Diego, CA, USA) Transcripts were aligned to the Human Feb. 2009 (GRCh37/hg19) assembly using TopHat (version 2.1) (46). Gene expression sets were prepared using ICount, which is based on HTSeq-count. Uniquely mapped reads were normalized to 10 million reads followed by log2 transformation. In order to avoid negative normalized values, 1 was added to each gene expression value. Data were analysed using Gene Set Enrichment Analysis software (47, 48) (University of California San Diego, San Diego, CA, USA) and the differentially expressed gene sets (nominal p-value < 0.05) displayed. The results of the gene set enrichment analysis are displayed in Supplementary Table. 2. The heat map was created using Heat mapper online tool, plotting the count per million per sample (49). The RNA-seq data were analysed as follow, transcripts with more than 2 counts in 3 or more of the samples were kept, for data normalization TMM (edgeR), weighted trimmed mean of M-values (to the reference) was used (50), the data were annotated using biomaRt, using Ensembl. The count data was transformed to log2-counts per million (logCPM) using voom, estimating the mean-variance relationship and the differential expression was assessed using a moderated t-test using the linear model framework from the limma package and the adjusted p-value calculated by using FDR, Benjamini-Hocheberg correction (Supplementary Table. 3). Differentially expressed genes (FDR adj. p-value < 0.05 and Log2 fold change of 0.5) were displayed in a volcano plot.
Western blot and nuclear fractionation
After washing with ice-cold phosphate-buffered saline (PBS), hCMEC/D3 cells were lysed with cell lysis buffer (Cell Signaling Technology, Boston, MA, USA) containing a protease and phosphatase inhibitor cocktail (Roche, Almere, The Netherlands, and Cell Signaling Technology, Boston, MA, USA, respectively) on ice, following the manufacturer’s instructions. Nuclear fractions were isolated using the NE-PER extraction kit (Thermo Fisher Scientific, Rockford, IL, USA), following the manufacturer's guidelines. All samples were diluted in sample buffer (BioRad Hercules, CA, USA) (65.8 mM Tris-HCl, pH 6.8, 2.1% SDS, 26.3% (w/v) glycerol, 0.01% bromophenol blue) and heated to 95°C for 3 min. For whole cell lysates, hCMEC/D3 were removed from the media and lysed in sample buffer. Lysates were separated on SDS-PAGE followed by transfer to nitrocellulose for immune-blot analysis. Blots were blocked for 1 h at room temperature with blocking buffer (Azure Biosystems, Inc, Sierra CT, Dublin, CA, USA). Subsequently, membranes were incubated in blocking buffer containing 0.1% Tween-20 with antibodies against claudin-5 (Santa Cruz, Dallas, TX, USA), SNAI2 (Abcam, Cambridge, United Kingdom) and GAPDH (Proteintech, Manchester, United Kingdom). Primary antibodies were detected and quantified by incubation with IRDye secondary antibodies (LI-COR) and use of Azure Sapphire Biomolecular Imager (Azure Biosystems, Inc, Sierra CT, Dublin, CA, USA).
Immunofluorescence microscopy
hCMEC/D3 cells were seeded in 8 well µ-slides (Ibidi, München, Germany) and treated as described in the cell culture section. Cells were fixed with 4%, 1.6% paraformaldehyde, or ice-cold methanol (Sigma-Aldrich, Saint Louis, MO, USA) and then permeabilized for 5 minutes using 0.05% Triton-X100 in PBS (Sigma-Aldrich, Saint Louis, MO, USA). Unspecific binding was prevented with 5% normal goat serum. Cells were then incubated with mouse anti-CLDN5 (Santa Cruz, Dallas, TX, USA), mouse anti-SNAI2 (Abcam, Cambridge, United Kingdom), mouse anti-CD31 (DAKO, Naestved, Denmark), Delta-4 Antibody (G-12) (Santa Cruz, Dallas, TX, USA) and rabbit anti-Zonulin-1 (ZO1) (Thermo Fisher Scientific, Rockford, IL, USA). Primary antibodies were visualized using goat anti-mouse Alexa 555/488 (Molecular Probes, Eugene, OR, USA). Nuclei were visualized using Hoechst (Molecular Probes, Eugene, OR, USA). Stainings were imaged using the Leica SP8 microscope (Leica, Mannheim, Germany) or LIPSI Ti2 (Nikon, Tokyo, Japan)
3D in vitro sprouting assay
Spheroids were generated for the sprouting assay. In brief, hCMEC/D3 and hiPSCs pericytes were resuspended in a ratio of 20:1 in EGM-2 medium containing 0.25% methylcellulose (4.000 cP, Sigma-Aldrich, Saint Louis, MO, USA). To form spheroids, the mixture of cells was seeded in a 24 well plate and flipped upside down. After 24h, the spheroids were collected and resuspended in 1,5 mg/ml collagen Type I rat tail mixture (Enzo science, Farmingdale, NY, USA) and plated in a 24 well plate upside down until complete polymerization. After 30 min, EGM-2 medium was administered and wells were incubated at 37°C and 20% O2, 5% CO2 for 5 days or at 1% O2, 5% CO2 for 5 days. Images were taken using the Nikon LIPSI Ti2 confocal spinning disk imaging system (Nikon, Tokyo, Japan), 10 x objective, and adjusted for brightness/contrast in ImageJ. Sprouting number and length were analysed using the ImageJ plugin NeuronJ .
Immunohistochemistry on post-mortem human brain tissue
Brain tissue from 5 patients with clinically diagnosed and neuropathologically confirmed capCAA, 6 AD and 6 non-demented control (NDC) cases without neurological diseases was obtained after autopsy (post-mortem delay < 8hr) and immediately frozen in liquid nitrogen (in collaboration with the Netherlands Brain Bank, Amsterdam). The Netherlands Brain Bank received permission from the ethical committee of the VU University Medical Center Amsterdam, the Netherlands to perform autopsies, for the use of the material and for access to medical records for research purposes. Cortical grey matter samples from the superior occipital gyrus (SOG) were selected and used for staining. All patients and controls, or there next of kin, had given informed consent for autopsy and use of their brain tissue for research purposes. Clinical data are presented in Supplementary Table. 4.
For immunohistochemical analysis, 5 µm thick cryosections of frozen brain tissues were fixed in ice-cold acetone for 10 min. After washing with PBS, sections were incubated overnight at 4°C with primary antibodies against SNAI2 (Abcam, Cambridge, United Kingdom). Subsequently, sections were washed with PBS and incubated with Envision Dual Link (DAKO, Glostrup, Denmark) for 1h at room temperature, followed by visualization with the peroxidase substrate 3,3′-diaminobenzidine (DAKO, Glostrup, Denmark). Sections were incubated with hematoxylin (Sigma-Aldrich, Saint Louis, MO, USA) for 1 min and thoroughly washed with tap water for 10 min. Ultimately, sections were dehydrated with ethanol followed by xylene (Sigma-Aldrich, Saint Louis, MO, USA) and mounted with Entellan (Merck, Darmstadt, Germany). Immunofluorescent labelling was performed as follows: after fixation in ice-cold acetone for 10 minutes, the sections were incubated for 30 min with 10% normal goat serum and 0.1% Triton X-100 (Sigma-Aldrich, Saint Louis, MO, USA) and afterwards incubated overnight at 4°C with antibodies against SNAI2 (Abcam, Cambridge, United Kingdom) or ANGPTL4 (Abcam, Cambridge, UK), and sections were stained with UEA-1 (Vector Lab, Burlingame, CA, USA). The primary antibodies were visualized by incubation with goat anti-mouse Alexa 555 (Molecular Probes, Eugene, OR, USA), donkey anti-rabbit Alexa 647 (Molecular Probes, Eugene, OR, USA) or Streptavidin 488 (Molecular Probes, Eugene, OR, USA) for 1 hour at RT. Next, to visualize Aβ aggregates, sections were incubated for 5 minutes with Thioflavin-S (Sigma-Aldrich, Saint Louis, MO, USA) and washed with ethanol afterwards. After washing with PBS, Hoechst (Molecular Probes, Eugene, OR, USA) was used for nuclear staining and slides were mounted in Mowiol (Sigma-Aldrich, Saint Louis, MO, USA).
Image acquisition and analysis
Images of the DAB-stained tissue were obtained using a DM6000 (Leica, Mannheim, Germany), 4 random regions of interest (ROIs) were collected per sample and the results presented as average staining intensity per section. Fluorescent images were obtained using an Olympus VS200 (Olympus, Tokyo, Japan) slide scanner or a SP8 confocal microscope (Leica, Mannheim, Germany). Five specific ROIs with a Z-stacks of 6 µm and a 60x magnification were recorded and the results are presented as average staining intensity per section. Image deconvolution and analysis were done using Huygens Professional 21.10 software (Scientific Volume Imaging B.V., Hilversum, The Netherlands) and NIS elements (version 5.30.03, Nikon Europe B.V., Amsterdam, The Netherlands) or ImageJ (U.S. National Institutes of Health, Bethesda, MD, USA).