Chemicals, buffers, and miscellaneous reagents
MLN4924/Pevonedistat (catalog #A-1139) was bought from Active Biochem, China. A 500 µM stock solution was prepared in 100% DMSO. CSN5i-3 was obtained from MedChem Express (catalog #HY-112134). A 1 mM stock solution was prepared in 100% DMSO. Miscellaneous reagents were purchased from Sigma Aldrich/Merck (Darmstadt, Germany), VWR International GmbH (Darmstadt, Germany), Carl Roth GmbH (Karlsruhe, Germany), and ThermoFisher Scientific, Netherlands) and were of the highest purity degree available.
Mice
Wildtype C57BL/6J mice were initially obtained from Charles River Laboratories (Sulzfeld, Germany). Cx3cr1EGFP/+ mice, which were originally obtained from the Jackson Laboratories (strain 005582; (52)), were established on a pure C57BL/6J background. Mice were housed under standardized light-dark cycles in a temperature-controlled air-conditioned environment under specific pathogen-free conditions at the Center for Stroke and Dementia Research (CSD), Munich, Germany, with free access to food and water. Animals were sacrificed under anaesthesia with a mixture of midazolam (5 mg/mL), medetomidine and fentanyl (MMF). Mouse maintenance and experiments were reviewed and overseen by the institutional animal use and care committee of the local authorities (Regierung von Oberbayern, ROB, Germany) and performed in accordance with the procedures provided by the animal protection representative of CSD.
Cell culture and cell lines
Cells were cultivated in a temperature- and humidity-controlled incubator at a temperature of 37°C and 5% CO2. Fetal calf serum (FCS) from an EU-approved origin was obtained from Invitrogen-ThermoFisher Scientific (Karlsruhe, Germany) and heat-inactivated prior to usage. Other cell culture reagents, media and supplements were also bought from Invitrogen-ThermoFisher Scientific.
Cell culture of BV2 microglia
The BV2 microglia cell line CVCL_0182 was initially obtained from Dr. Markus Kipp (formerly RWTH Aachen University, Germany; now University of Rostock, Germany), who obtained the cells from Interlab Cell Line Collection (ICLC), Italy (accession number: ICLC ATL03001; http://wwwsql.iclc.it/det_list.php). The BV2 cell line was cultured in RPMI1640 (Roswell Park Memorial Institute 1640) GlutaMAX medium containing 10% fetal bovine serum (FBS), 1% penicillin/streptomycin (P/S), and maintained in poly-L-ornithine (0.01%)-coated T75 flasks (Sigma Aldrich, Taufkirchen, Germany) in a humidified incubator at 5% CO2 and 37°C. Cells were split every 2–3 days until passage 20. For inflammatory stimulation experiments, BV2 cells were stimulated with 20 ng/mL or 100 ng/mL mouse TNF-α (product # 300-01A, Peprotech, Hamburg, Germany) as indicated. For CSN5 mimicry or inhibition experiments, cells were pre-treated with 500 nM MLN4924 and 1 or 4 µM CSN5i-3, respectively, for 2–4 h in all experiments. Final DMSO concentrations in the vehicle groups were between 0.1% and 0.4% and did not interfere with cell viability as verified in scouting experiments.
Human cerebral microvascular endothelial cell culture (hCMEC/D3)
The immortalized hCMEC/D3 cell line was seeded on rat tail collagen type I (product #08115; Merck Millipore, Darmstadt, Germany) coated T-25 flask in EndoGRO-MV Complete Media Kit (product #SCME004; Merck Millipore), and maintained at 5%CO2 and 37°C exposure. Cells were split when they reached a confluent monolayer. For stimulation experiments, hCMEC/D3 cells were treated with 100 ng/mL human TNF-α (product #300-01A, Peprotech) as indicated. Pre-treatment with MLN4924 and CSN5i-3 was performed essentially as described above for BV2 cells (500 nM MLN4924 for 2 h, 1 µM CSN5i-3 for 4 h; final concentration of 0.1% and 0.1% DMSO in vehicle controls).
Primary cell cultures
Primary neuronal culture
Primary neurons were derived from p0-p1 wildtype C57BL/6J neonatal mouse pups. Mice were decapitated by scissors and the heads immediately placed in ice-cold dissection buffer (97.5% HBSS Ca2+ and Mg2+ free, 110 µg/mL sodium pyruvate, 0.1% glucose, 10mM HEPES, pH 7.3). Brain dissection was performed in the ice-cold dissection buffer in a 6 cm petri-dish under a Zeiss Stemi 305 microscope (Zeiss, Oberkochen, Germany). Cortical tissues were separated from the brain and the hippocampi removed. The cortex was transferred using a fire-polished glass Pasteur pipette into pre-warmed 2 mg/mL papain buffer at 37°C for 15 min. The cell pellet was washed in cold plating medium (86.55% MEM Eagle’s with Earle’s BSS, 10% FBS, 0.45% glucose, 1 mM sodium pyruvate, 2 mM glutamine, 1% P/S once and triturated in pre-warmed plating medium using a pipette. After trituration, the total cell suspension was filtered through a 40 µm filter and cells plated on coverslips or dishes, coated with poly-L-Lysine (0.05 mg/mL). The plates were incubated at 37°C in a humidified incubator with 5% CO2 and the medium replaced by growth medium (96% neurobasal medium, 2% B27, 2 mM glutamine, 1% P/S) the next day. Half of the growing medium was then changed twice a week for 10 to 14 days before any treatment. Neuronal cultures were pre-treated with MLN4924, CSN5i-3, or solvent control for two hours before exposure to OGD.
Primary microglia culture and mixed brain culture
Primary microglia were isolated from the cerebral cortex of p0-p2 wildtype C57BL/6J neonatal mouse pups and digested with papain following positive selection of CD11b + microglia/myeloid cells by CD11b magnetic bead enrichment (Miltenyi Biotec, Bergisch Gladbach, Germany). Isolated microglia were seeded in plates and grown in DMEM/F12 medium supplemented with 10% FCS, 1% P/S and 10 ng/mL GM-CSF. After 14 days, microglia were subjected to the indicated treatments.
CX3CR1EGFP/+ microglia migration was examined in mixed brain cultures. Brain cortices were isolated from CX3CR1EGFP/+ p0-p2 pups in Hank’s Balanced Salt Solution (HBS) supplemented with 8% NaHCO3 and 1 µM HEPES. Tissues were digested in papain solution and all cells plated in normal media (MEM without phenol red, 20% glucose, 8% NaHCO3, 0.1 mg/mL transferrin) with 0.5% P/S, 10% FCS, 2 mM L-glutamine, 0.025 mg/mL insulin in a 96-well cell imaging plate (product #0030741030; Eppendorf, Hamburg, Germany). Media were replaced on the second day by normal media and changed twice a week for two weeks.
For inflammatory stimulation experiments of primary microglia cells/cultures, see ‘cell culture of BV2 microglia’.
Transfection of BV2 microglia cells with siPool
BV2 cells were cultivated in the RPMI medium as described previously (53). Specific siRNAs for the mouse Cops5 gene were designed by siTOOL Biotech (Planegg, Germany). Transfection of BV2 cells was accomplished using Lipofectamine RNAiMax (Invitrogen-ThermoFisher Scientific) in Opti-MEM medium (catalog #31985070, Gibco-Invitrogen- ThermoFisher Scientific) according to the manufacturer’s protocol. Treatment of cells was started 48 h after transfection and was performed as indicated in Results.
Proteomics analysis
BV2 cells were cultured in full medium in 10 cm cell culture dishes until they were confluent. MLN4924 (500 nM), CSN5i-3 (1 µM), or solvent control (0.01% DMSO) were added and the culturing continued for 6 h. After the incubation, cells were washed once with PBS, removed from the cell culture plate with a scraper, collected in tubes, and centrifuged in 1.5 ml Eppendorf tubes for 3 min to remove remaining PBS buffer, snap-frozen, and stored at -80°C until further processing.
Thereafter cells were collected and lysed in urea buffer (9 M Urea, 50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 1x Roche protease inhibitor cocktail) followed by short sonification. Samples were cleared by centrifugation and protein amounts were adapted. Protein reduction was performed with dithiothreitol (DTT; 5 mM final) for 25 min at 56°C and protein alkylation by the addition of iodoacetamide (14 mM final) for 30 min at room temperature. Protein mixtures were quenched with DTT and diluted 1:5 with 1 M Tris-HCl, pH 8.2. For increased peptide recovery, proteins were digested at room temperature for 3 h with LysC (FUJIFILM, 2 µL/100 µg protein) before overnight tryptic digest at 37°C (0.5 µg/100µg protein). The following day, digestion was stopped with 10% trifluoroacetate (TFA). To increase analysis depth, samples were pre-fractioned by a C18-SCX custom-made stage tip (54). Fractions were eluted stepwise with increasing NH4AcO concentrations (20 mM to 500 mM) and desalted on a separate C18 stage tip. Desalted peptides were loaded on a custom-made 75 mm x 15 cm fused silica capillary filled with C18-AQ resin (Reprosil Pur 120 HPLC column, 1.9 µm, Dr. Maisch HPLC GmbH, Ammerbuch, Germany) using an Easy-nLC1200 liquid chromatography. Samples were separated for 140 min with a 2.4–80% acetonitrile gradient in 0.1% formic acid using a Q Exactive HF mass spectrometer (MS; ThermoFisher Scientific). MS raw data were processed with MaxQuant (version 1.6.0.1) and loaded into Perseus (version 1.6.5.0), where matches to common contaminants, reverse identifications, identifications based only on site-specific modifications and with less than 2 peptides and MS/MS counts were removed. Only proteins with LFQ intensities in 3 out of 4 biological replicates in at least one experimental group were kept for the subsequent label-free quantification (LFQ). LFQ intensities were log2 transformed and missing values were replaced with random numbers drawn from a normal distribution. Student’s t-tests were used to determine the statistical significance of the abundance alterations of proteins detected between control and treated conditions. Proteins with P < 0.05, FDR < 0.05 and t-test difference > 0.5 or <-0.5 were considered to be significantly increased or decreased, respectively. Functional annotations were performed using DAVID (https://david.ncifcrf.gov/).
RNA isolation and quantitative real-time PCR (RT-qPCR)
Total RNA was extracted from cells using TRIzol (catalog #15596018, ThermoFisher Scientific). RNA was reverse-transcribed into cDNA using ReverAid First Strand cDNA Synthesis Kit (catalog #K1622, ThermoFisher Scientific). Quantitative RT-PCR was performed using a RotorGene thermocycler (Qiagen Corbett, Hilden, Germany) and cDNA quantified with SYBR green. Primers used were: i) mouse forward and reverse primers of - Ccl2, 5’-CATCCACGTGTTGGCTCA-3’, 5’-GATCATCTTGCTGGTGAATGAGT-3’: Il12-β, 5’-TTCTCACCGTGCACATCC-3’, 5’-GACCGGCACTGAGAGGAG-3’; β-actin, 5’-GGAGGGGGTTGAGGTGTT-3’, 5’-GTGTGCACTTTTATTGGTCTCAA-3’; Tnf-α, 5’-CATCTTCTCAAAATTCGAGTGACAA-3’, 5’-TGGGAGTAGACA AGGTACAACCC-3’; iNos, 5’-GGAGCCTTTAGACCTCAACAGA-3‘, 5‘-AAGGTGAGCTGAACGAGGAG-3‘; ii) human forward and reverse primers of - GAPDH, 5’-AAGGTGAAGGTCGGAGTCAA-3’, 5’-AATGAAGGGGTCATTGATGG-3’; ICAM1, 5’-CCTTCCTCACCGTGTACTGG-3’, 5’-AGCGTAGGGTAAGGTTCTTGC-3’; VCAM1, 5’-TGCACAGTGACTTGTGGACAT-3’, 5’-CCACTCATCTCGATTTCTGGA-3’; E-selectin, 5’-GAGTGCACATCTCAGGGACA-3’, 5’-ACTGCCAGGCTTGAACATTT-3’; CCL2, 5’-AGTCTCTGCCGCCCTTCT-3’, 5’-GTGACTGGGGCATTGATTG-3’, respectively. The cycle time (Ct) values were normalized with β-actin and evaluated by the ΔΔCt method.
Western blotting
Cells were lysed in NuPAGE-LDS lysis buffer containing 125 mM DTT. Protein fractions were separated by 11% or 15% SDS-PAGE and blotted onto a nitrocellulose membrane. A 3% bovine serum albumin (BSA) solution was used for blocking and the following primary antibodies were applied for target protein detection: anti-NF-kB IkB-alpha (catalog #9242S, Cell Signaling Technologies, Frankfurt am Main, Germany), anti-NF-kB pI-kB-alpha (catalog #9246S, Cell Signaling), anti-cullin1 (catalog #sc-17775, Santa Cruz, Heidelberg, Germany), anti-NEDD8 (cataog #2745S, Cell Signaling), anti-VE-Cadherin (catalog #sc-9989, Santa Cruz), anti-occludin (cataog #71-1500, ThermoFisher Scientific), anti-Jab1 (catalog # sc-13157, Santa Cruz), anti-CSN8 (catalog #PW8290, Enzo Life Sciences GmbH, Lörrach, Germany), anti-Akt (catalog #9272S Cell Signaling), anti-p-Akt (catalog #9275 Cell Signaling), anti-claudin-5 (catalog #341600, ThermoFisher Scientific), anti-β-actin (catalog #sc-47778, Santa Cruz). As secondary antibodies, HRP-conjugated anti-mouse (catalog #ab6820, Abcam, UK) or anti-rabbit antibodies (catalog #BYT-ORB43514, Biozol, Eching, Germany) were used. Blots were developed with SuperSignal West Dura Extended Duration Substrate (catalog #34076 ThermoFisher Scientific) and visualized applying an Odyssey® Fc imager (LiCOR, Hamburg, Germany) and quantified by ImageJ-FIJI version 2.0.0.
Phagocytosis assay
BV2 cells and primary microglia were cultured into 24-well plates on coverslips at a density of 5x104 and 1x105 cells per well, respectively. Cells were pre-treated with MLN4924 or vehicle control for 2 h in serum-free medium. Cells then were incubated with or without TNF-α containing 0.05% fluorescently-labeled latex beads (catalog # L3030, Sigma-Aldrich, Taufkirchen, Germany) for 6 or 24 h. After incubation, cells were washed 5 times with ice-cold PBS, followed by fixation using 4% PFA for 15 min in the dark, and permeabilized with 0.2% Triton X-100 for 10 min. The slides were mounted with the mounting medium containing DAPI (catalog #H1200-10, Vectashield, Eching, Germany), and the phagocytosis rate scored by fluorescence microscopy on a Leica Dmi8 microscope. Phagocytosing cells were defined as follows: phagocytosing cells (%) = number of cells that contained latex beads/number of total cells as scored by DAPI-positivity.
Transwell permeability assay
For the Transwell permeability assay, hCMEC/D3 cells were seeded at a density of 2x105 cells/well in 300 µL endothelial medium (ENdoGRO-MV Complete Culture Media Kit SCME004, Merck Millipore) on the membranes of the Transwell inserts (6.5 mm Transwell-COL collagen-coated 0.4 µm Pore PTFE membrane insert 3495, Corning (Merck). Following treatment with MLN4924, CSN5i-3, or vehicle and/or OGD/RO (see above), 5 µM Lucifer yellow was added to the upper chamber of the Transwell insert and the device incubated for 60 min (5% CO2, 37°C). For quantification, 100 µL of medium from the lower chamber was transferred to a black 96-well polystyrene plate and the Lucifer yellow signal measured at 530 nm (excitation at 485nm) with a fluorescence microplate reader (Perkin Elmer Enspire). The apparent permeability (Papp) was calculated by the following equation: Papp = (dQ / dt) / (A x C), where dQ / dt is the amount of drug (here Lucifer yellow) transport within a given time period; A is the surface area of the insert; and C is the initial concentration of the drug in the upper chamber at time 0 h.
Migration of microglia
Mixed brain cells were seeded in a 96-well cell imaging plate (catalog #0030741030, Eppendorf) in Hibernate-A medium (catalog #A1247501, ThermoFisher Scientific), pre-treated with MLN4924 or vehicle control and stimulated with or without CXCL12 at a concentration of 100 ng/mL. Cells were monitored under the FITC-channel of a fluorescence microscope (Leica Dmi8) every 5 min for 14 h applying the live-imaging modality. Tracking analysis of cells was performed using the plugin ‘Manual Tracking’ from the ImageJ software package (National Institutes of Health, NIH, Bethesda, USA). Migration was further analyzed with the ‘Chemotaxis and Migration Tool’ software from Leica to assess the accumulated distance as a parameter of migration response.
Organotypic brain slice cultures
Organotypic brain slice cultures were prepared as described before (55, 56) with modifications. Brains were removed from p5-8 neonatal wildtype C57BL/6J or Cx3cr1EGFP/+ mice by decapitation according to animal handling laws. Hippocampi and neocortices were dissected and, as depicted in Fig. 8, 350 µm-thick sagittal sections were cut from the hippocampal and cortical tissue, using a Mcllwain tissue chopper (Model TC752, Mickle Laboratory Engineering Company, Goose Green, UK). Intact sections were carefully selected under a Zeiss Stemi 305 dissection microscope in dissection medium (MEM, catalog #32360-026 (Gibco-Invitrogen-ThermoFisher Scientific), containing 1% P/S and 10 mM Tris HCl, pH 7.2). Slices were incubated in cold dissection medium for 30 min before plating, and two slices were plated onto an polytetrafluoroethylene (PTFE) membrane insert (0.4 µm, 30 mm diameter, PICMORG50 from Merck-Millipore). Slice culture medium containing 50% HEPES-buffered Minimum Essential Medium (MEM) (catalog #32360026), 25% heat-inactivated horse serum (catalog #26050088, Merck-Sigma), 25% HBSS (catalog #14025050) and 1 mM L-glutamine (catalog #25030081) (all but horse serum from Gibco-Invitrogen-ThermoFisher Scientific) was changed one day after initial seeding and subsequently twice a week. Treatment was applied directly to the slice culture medium. Stock solutions of MLN4924 or CSN5i-3 were prepared by dissolving compounds in 100% DMSO at a concentration of 500 µM and 1 mM. Slice cultures were pre-treated with 10 µM MLN4924 or CSN5i-3 for 24 h (final DMSO concentration 2% and 1%, respectively) before further being exposed to oxygen-glucose deprivation (OGD) stress (see below).
Oxygen-glucose deprivation (OGD) and reoxygenation (RO)
Oxygen glucose deprivation (OGD) was performed on day 10–14 of the primary neuronal cultures, on fully confluent monolayers of hCMEC/D3 cells, and on 14-day-old organotypic brain slice cultures. Following prior culture in glucose-containing medium, the cell culture medium was changed to DMEM without glucose, glutamine, and phenol red (catalog #A1443001, Gibco-Invitrogen-ThermoFisher Scientific), and flushed with a gas-mix of 95% N2 and 5% CO2 for 10 min before placing the cells/organotypic slices into a humidified hypoxia chamber (Hypoxie Glove Box HGB-090-1, Toepffer Lab Systems, Adelberg, Germany; 95% N2, 5% CO2, 1% O2, 45% humidity, 37°C) for indicated time intervals. OGD was terminated by returning the cells/organotypic slices to normoxic conditions with glucose-containing medium (RO, reoxygenation).
Cell Counting Kit-8 (CCK-8) viability assay
Primary neuronal cells were seeded at the same density in 96-well plates and incubated with 100 µL growth medium for 10–14 days. The cells were then treated with MLN4924, CSN5i-3, or vehicle control, followed by OGD stress for various time intervals as indicated. Cell viability was determined after a 24 h re-oxygenation period using the CCK-8 kit (catalog #96992, Sigma- Aldrich). Briefly, to each well of the 96-well plate, 10 µL CCK-8 reagent was added to the medium and cells incubated for 2 h at 37°C. The spectrophotometric absorbance of each well was determined using a multi-label microplate reader (Enspire, Perkin Elmer LAS GmbH, Rodgau, Germany) at a wavelength of 450 nm.
Evaluation of cell damage in organotypic brain slice cultures
Propidium iodide (PI, catalog # 81845, Sigma-Aldrich) was used to label damaged cells in the organotypic slice cultures. After pre-treatment with MLN4924 or CSN5i-3 and OGD/RO, brain slices were incubated with 7 µM PI for 15 min at 37°C. Afterwards, images were directly acquired using a Leica Dmi8 microscope (Leica Microsystems GmbH, Wetzlar, Germany) with a 4x dry objective. Randomly-chosen PI-positive areas were manually quantified and normalized to the respective total tissue area, using Image J-Fiji (version 2.0.0). For the pharmacological blockade experiments with anti-TNF drugs, the investigator scoring the PI-positive areas was blinded for group assignments.
Microglia morphology changes in brain slice cultures and in vivo
In order to investigate microglia phenotypes, brain slice cultures were prepared from Cx3cr1EGFP/+ mice. Following pre-treatment with MLN4924 or CSN5i-3 and OGD/RO (for details, see above), representative pictures from brain slices at a 14-day in vitro (DIV) period were acquired before and after fixation using a 4x or 20x dry objective (Leica Dmi8 fluorescence microscope, Leica Microsystems GmbH) or 40x oil immersion objective (Zeiss AiryScan 880 confocal microscope).
Immunofluorescence staining of cells and organotypic brain slices
BV2 microglial cells, primary neuronal cells and hCEMC/D3 cells were cultured on coverslips in 24-well plates. Following the above-described treatments, cells were fixed with 4% paraformaldehyde (PFA) for 15 min in the dark, permeabilized with 0.2% Triton X-100 for 10 min, blocked with SuperBlock solution (catalog #37515 ThermoFisher Scientific) for 1 h, and incubated with primary antibody overnight at 4°C using the following antibodies: anti-NF-kB p65 (catalog #8242, Cell Signaling, 1:500), anti-beta-tubulin III (catalog #T8578, Sigma-Aldrich), anti-VE-cadherin (catalog #F8 sc-9989, Santa Cruz Biotechnology, 1:100). After three washes with PBS, slides were incubated with secondary antibody (goat anti-rabbit conjugated to Alexa Fluor 555, 1:1000 or goat anti-mouse conjugated to Alexa Fluor 647, 1:1000) for 1 h, counter-stained with DAPI, and coverslips finally fixed, and sealed on the slides. Images were acquired with a fluorescence microscope (Leica Dmi8) or a confocal microscope (Zeiss 880 AiryScan).
Brain slices were fixed in 4% PFA for 1 h at room temperature and permeabilized in PBS containing 0.5% Triton X-100 (PBS-T) for 30 min. Afterwards, slices were cut with the membrane from the insert using forceps and placed into a wet chamber, followed by blocking with PBS containing 0.5% Triton X-100 and 5% normal goat serum (catalog #ab7481, Abcam). To stain for neurons, slices were subsequently incubated with the primary antibody NeuN (catalog #26795-1-AP, Proteintech, UK) at 1:500 dilution in blocking solution overnight at room temperature. After three 10 min washes with PBS-T, slices were incubated with secondary antibody (goat anti-rabbit antibody conjugated to Alexa Fluor 647, 1:250, for 3 to 5 h at room temperature in the dark). After three 10 min washes with PBS-T, slices were circled with white filling paste and mounted with Fluoromount aqueous mounting medium (catalog #F4680, Sigma-Aldrich) and analyzed with a fluorescence microscope (Leica Dmi8). NeuN-positive areas were manually quantified and normalized to the respective total tissue area using Image J-Fiji (version 2.0.0)
Statistical analysis
Statistical analysis was performed using GraphPad Prism Version 8 software. Unless stated otherwise, data are represented as means ± standard deviation (SD). After testing for normality by Shapiro-Wilk test, data were analyzed either by two-tailed Student’s T-test, Mann-Whitney U test, ordinary one-way ANOVA as appropriate, or two-way ANOVA. To account for multiple comparisons, either Dunnett’s or Bonferroni posthoc multiple comparisons tests were applied as appropriate. For information on the statistical analysis of the proteomics/mass spectrometry data, see the dedicated paragraph ‘proteomics analysis’ above. Differences with P < 0.05 were considered to be statistically significant.