Neuronal cultures Mouse primary hippocampal neurons were obtained from postnatal day 0 C57BL/6 and FVB mice and prepared as previously described 78,85. Housing and euthanasia were performed in compliance with the Canadian Council on Animal Care. Neurons used in imaging experiments were cultured at low density (50,000 neurons per 35 mm (14 mm glass) dish (MatTek, # P35G-1.5-14-C)). Neurons used in RNA-seq and Simple Western experiments were cultured at 300,000 neurons per well in 100 µm Transwell membranes (Thermo Fisher Scientific, # 35102) in 6-well dishes 80.
Dissociated spinal cord cultures from E13 CD1 mice were prepared as previously described 96. Cells were plated on poly-D-lysine (Sigma, #P6407) - and Matrigel-coated glass coverslips in 6-well dishes. The culture medium was as described 96 with the addition of 1% B27 (Gibco Life Technologies, Burlington, ON, Canada, #17504044), 0.032 g/mL sodium bicarbonate and 0.1 g/mL dextrose. Cultures were maintained for at least 3 weeks to ensure motor neuron maturation.
Human motor neurons were differentiated from iPSCs as previously described 117,118. CV-B (wild type) iPSCs were a gift from the Zhang Lab 132 and HNRNPA2B1 D290V-1.1 and − 1.2 human iPSCs were generated in the Yeo lab 118. Human iPSCs were grown on Matrigel-coated 10 cm tissue culture plates. When cells were 80–90% confluent, they were split into 6-well plates at 1×106 cells/well in 1× N2B27 medium (DMEM/F12 + Glutamax, 1:200 N2 supplement, 1:100 B27 supplement, 150 mM ascorbic acid, and 1% Penicillin/Streptomycin) supplemented with 1 µM dorsomorphin (Tocris, #3093), 10 µM SB431542 (Tocris, #1614), 4 µM CHIR99021 (Tocris, #4423) and 10 µM Y-27632 hydrochloride (ROCK inhibitor; Tocris, #1254). The seeding day was counted as day 1. On days 1–5, the cells were refed daily with the same medium as on day 1, but with the ROCK inhibitor reduced to 5 µM. On days 7–17, the cells were refed daily with 1× N2B27 medium supplemented with 1 µM dorsomorphin, 10 µM SB431542, 1.5 µM retinoic acid (Sigma, #R2625), 200 nM Smoothened Agonist, SAG (EMD Millipore, #566660), and 5 µM ROCK inhibitor. On day 18, the cells were either plated on laminin-coated 10 cm plates at 1.2×107 cells per plate for continued differentiation or expanded in motor neuron progenitor MNP medium (1× N2B27 medium supplemented with 3 mM CHIR99021, 2 mM DMH1 (Tocris, #4126), 2 mM SB431542, 0.1 mM retinoic acid, 0.5 mM purmorphamine (Tocris, #4551), and 0.5 mM valproic acid (Tocris, #2815)) on Matrigel-coated plates. To expand motor neuron progenitors, cells were refed every other day with MNP medium. Laminin plates were prepared by serially coating them with 0.001% (0.01 mg/mL) poly-D-lysine (Sigma, #P6407) and poly-L-ornithine (Sigma, #P3655) followed by 20 µg/mL laminin (Life Technologies, #23017015). Cells were refed on day 18 and day 20 with MN medium (1 × N2B27 medium supplemented with 2 ng/mL glial cell-derived neurotrophic factor, 2 ng/mL bone-derived neurotrophic factor, and 2 ng/mL ciliary neurotrophic factor (all from R&D Systems, #212-GD, #248-BD, and #257-CF, respectively) supplemented with 1.5 µM retinoic acid, 200 nM SAG, and either 10 µM ROCK inhibitor on day 18 or 2 µM ROCK inhibitor on day 20. On days 22 and 24, cells were fed with MN medium supplemented with 2 µM DAPT and 2 µM ROCK inhibitor. On day 25, cells were split onto laminin-coated glass coverslips in a 12-well plate at 6.7x106 cells/ well in MN medium supplemented with 10 µM ROCK inhibitor. On day 27, cells were fed with MN medium supplemented with 2 µM ROCK inhibitor. On day 29, cells were stressed with 10 µM MG132 (Sigma, # M7449) for 7 h at 37°C. Cells were then fixed in 4% paraformaldehyde in phosphate-buffered saline and5 mM MgCl2 (PBSM) for 1 h at room temperature, washed once with 0.1 M glycine in PBSM for 10 min, and stored in PBSM at 4°C for IF staining and mRNA FISH.
Neuronal manipulation Neurons were stressed via 10 µM MG132 for the indicated times, hypoxia-reoxygenation (1% O2 for 3 h and 4 h recovery at 5% O2) using a hypoxia glove box (BioSpherix Xvivo System Model X3), or incubation with oligomers made from 500 nM amyloid-β (1–42) monomers (rPeptide, #1163-1) 133. As mature neurons cannot be transfected, plasmids were introduced into primary cultured mouse motor neurons by intranuclear microinjection. The injectate (the plasmid in 50% Tris-ethylenediaminetetraacetic acid (EDTA), pH 7.2) was clarified by centrifugation prior to insertion into 1 mm diameter quick-fill glass capillaries (World Precision Instruments) pulled to fine tips using a Narishige PC-10 puller (Narishige International USA, Inc., NY, USA). Cultures on coverslips were bathed in Eagle’s minimum essential medium without bicarbonate, supplemented with 5 g/L glucose, and adjusted to pH 7.4 in 35 mm culture dishes on the stage of a Zeiss Axiovert 35 microscope (Carl Zeiss Microscopy, LLC, USA) and microinjected using a Transjector 5246 or a FemtoJet Transjector and a Micromanipulator 5171 (all from Eppendorf, Hamburg, Germany). Following microinjection, coverslips were placed in regular culture medium containing 0.75% Gentamicin (Gibco) and maintained at 37°C in a 5% CO2 environment until analysis.
Plasmid transfection and analysis Plasmids expressing shRNAs in a lentiviral backbone were obtained through the McGill University library (https://www.sidonghuanglab.com/pooled-screening-libraries/service-request/) (Table S2). They were transfected by calcium phosphate into 293T cells and knockdown efficiency was tested 72 h later by western blotting (Table S2).
IF and smFISH Detailed protocols for these methods have been previously described 78. RNA FISH probes were designed using the Stellaris Probe Designer (LGC Biosearch Technologies; masking level: 5, oligo length: 20, minimum spacing: 2) (Table S2).
Image acquisition and analysis Images were acquired using a Nikon eclipse Ti-2 inverted widefield microscope equipped with a SPECTRA X Light Engine (Lumencor) and an Orca-Fusion Digital CMOS Camera (Hamamatsu) controlled by NIS-Elements Imaging Software. A 60× 1.40 NA oil immersion Plan Apochromat objective lens (Nikon) was used with a xy pixel size of 107.5 nm and a z-step of 200 nm. Chromatic aberrations were measured before imaging using 100 nm TetraSpeck™ Fluorescent Microspheres (Invitrogen, #T14792) and considered in the downstream pipeline.
Single mRNAs, peptides, and postsynaptic densities were identified with the MATLAB version of the FISH-quant (v3) 86. Post-detection analyses of subcellular mRNA distributions in neurons and simulations were performed with the second version of ARLIN 88. The code for ARLIN v1.0 and ARLIN v2.0 can be found here: https://github.com/LR-MVU/neuron. See the corresponding documentation for a detailed explanation on ARLIN’s functionalities. Briefly, in ARLIN v2.0, simulations were improved by mimicking the distributions of real mRNAs when selecting simulated mRNAs. To do this, the dendrite was divided into “bins” of 25 µm. The program first counts x real mRNAs found in the first bin of the dendrite (i.e., 0–25 µm from the soma). Then, the program selects x “simulated mRNAs” (i.e., randomly selected pixels) from only the first bin of the dendrite. This ensures that the concentration of simulated mRNAs near the soma matches the concentration of real mRNAs, but with random distributions within the bin. The program counts the number of real mRNAs found in each bin, then randomly selects that number of pixels within it as “simulated mRNAs”. With this improved simulation, the statistical likelihood of an mRNA localizing to the synapses or to another mRNA can be calculated. statistics are calculated for the localization of mRNAs to synapses or to another mRNA. The simulation is repeated 100 times and the localization statistics are averaged. This provides a more accurate comparison between random and biologically driven colocalization patterns than in the first version of ARLIN.
To quantify translation efficiency, cell and dendrite segmentations were performed manually using the “Define outlines” tool in FISH-quant. The smFISH and peptide spots in the cells were fit to a 3D Gaussian model based on the point spread function and the analysis was run in batch mode. The x, y, and z coordinates of the mRNAs and peptides in cells were exported as tabulated text files (.txt) recording the identity of each cell in each image file analyzed. We designed a Python pipeline to first calculate the number of nascent peptides in spots with a signal brighter to that corresponding to the the average intensity of one peptide. Secondly, the pipeline assigns each peptide to the closest mRNA in the cell. If the distance between an mRNA and its closest peptide exceeded the threshold (200 nm plus the chromatic aberration), then we considered it a non-translating mRNA. To remove repeated mRNAs and peptides within the threshold, we selected the brightest (i.e., brightest) peptide signal, and then the closest to a single mRNA. The percentage of translating mRNA was calculated by dividing the number of translating mRNAs by the total number of mRNAs per cell (https://github.com/LR-MVU/neuron).
We used Fiji (ImageJ 2.14.0, Java 1.9.0_322) to calculate the granularity of the FLUC-GFP signal. First, we generated a maximum projection for the GFP channel. Then, we outlined a region of interest to define each dendrite in a neuron and measured its area, mean fluorescent signal, and standard deviation (SD). The coefficient of signal variation—the SD divided by the mean—was used as a readout for GFP.
RNA extraction and RNA-seq After 17 days in culture134, primary hippocampal neurons were washed with PBS, and the somas were scraped from the membrane and placed into a tube. Somas were centrifuged for 2 min at 2,000 × g and resuspended in 400 µL ice-cold PBS. The somas were divided into two tubes, and 750 µL of Zymo RNA Lysis Buffer (ZymoResearch, # R1013) was added to each. To harvest the neurites, membranes were cut from the Transwell, placed face down in a 6 cm plate containing 750 µL of Zymo RNA Lysis Buffer, and incubated for 15 min on ice while tilting the plate every few minutes. The solution was transferred into an Eppendorf tube. RNA was isolated using a Zyma Quick RNA Miniprep Kit (ZymoResearch, #R1054). RNA library generation and Illumina sequencing was performed by the University of Montreal Genomic Platform. PolyA capture, Nextseq High Output paired-end run (2 × 75 bp, coverage ~ 50M paired-ends per sample). The raw data have been deposited in the Gene Expression Omnibus under accession number GSE202202.
RNA-seq analysis was performed on usegalaxy.org. Adaptors and reads with a quality below 20 within 4-base sliding windows were removed using Trimmomatic (galaxy version 0.38.0; https://doi.org/10.1093/bioinformatics/btu170). Trimmed single-end reads were aligned to the mouse mm10 genome using STAR (galaxy version 2.7.8a + galaxy0; https://doi.org/10.1093/bioinformatics/bts635) with default parameters, and the number of reads per transcript was determined using featureCounts (galaxy version 2.0.1 + galaxy2; https://doi.org/10.1093/bioinformatics/btt656) using default parameters. Differential gene expression was determined using DESeq2 (galaxy version 2.11.40.7 + galaxy1; https://doi.org/10.1186/s13059-014-0550-8) using default parameters. Gene ontology analysis to identify biological processes enriched in differentially expressed genes was performed using geneontology.org.
To validate the data by RT-qPCR, 25 ng of RNA isolated from the soma or neurites was reverse transcribed into cDNA using iScript™ Reverse Transcription Supermix (Bio-Rad) following the manufacturer’s instructions. For qPCR, cDNAs were diluted two-fold in water. PCR was performed in 5 µL reactions consisting of 1 µL DNA, 2.5 µL PowerUp SYBR Green Master Mix (Thermo Fisher Scientific) and 0.25 µL of each primer (at 1 µM) on a Viaa 7 Real-Time PCR System (Thermo Fisher Scientific; 45 cycles). Standard curves were generated using a log titration of N2A genomic DNA (50–0.05 ng) and used to quantify the cDNA. The primers used are listed below.
Simple Westerns Primary hippocampal neurons grown on Transwell membranes were treated with 10 µM MG132 for 7 h at 37°C. After washing with ice-cold PBS, somas were first scrapped from the upper part of the membrane and then neurites were scraped from the bottom membrane and resuspended in ice-cold PBS. After centrifugation, somas and neurites were resuspended in radioimmunoprecipitation buffer (150 mM NaCl, 10 mM Tris-HCl pH 7.5, 0.1% sodium dodecyl sulfate (SDS), 1% Triton X-100, 1% sodium deoxycholate, and 5 mM EDTA pH 8.0). Protein extracts from somas and neurites were stored at -80°C. Simple Westerns (Bio-Techne) were performed in multiplex on a Jess automated western blot system (Bio-Techne) following the manufacturer’s instructions (Table S2).
Identification of RBPs binding to the mouse HSPA8 3′ UTR PP7-HSPA8 and PP7-LacZ RNA were first PCR amplified from N2A genomic DNA or a plasmid (donated by Dr. Jerry Pelletier) using the primers listed below, and then in vitro transcribed with a MEGAshortscript™ T7 Transcription Kit (Invitrogen, # AM1354) following the manufacturer’s instructions. After transcription, RNA was treated with 2 units of Turbo DNase and then purified by phenol-chloroform extraction and ethanol precipitation. RNA was resuspended in 10 mM Tris containing 0.2 U/mL RNaseOUT. Small samples were resolved on a 0.5X TBE agarose gel and their A260/A280 ratios were measured using a nanodrop to verify the purity of the RNAs. PP7-HSPA8 and PP7-LacZ RNA were heated for 2 min at 95°C, allowed to cool to room temperature to allow PP7 loops to form, and stored at -80°C.
To prepare crude N2A extracts, cells were differentiated into the neuronal phenotype for 3 days: one day in DMEM supplemented with 5% fetal bovine serum (FBS) and 20 µM retinoic acid, one day in 2.5% FBS and 20 µM retinoic acid, and one day in 1.25% FBS and 20 µM retinoic acid. Half of the cells were treated with 10 µM MG132 for 7 h at 37°C. Cells were washed once with ice-cold 1× PBS and pelleted by centrifugation. The cell pellets were washed once with 1× PBS and 1 mM phenylmethylsulfonyl fluoride. The supernatant was removed and cells were stored at -80°C. The pellets were thawed on ice, resuspended in three volumes of N2A lysis buffer (50 mM Tris-HCl pH 7.5, 100 mM NaCl, 1 mM MgCl2, 0.1 mM CaCl2, 1% IGEPAL CA-360, 0.5% deoxycholic acid, 0.1% SDS, 1 mM phenylmethylsulfonyl fluoride, 1 mM dithiothreitol, 1× Complete Protease Inhibitor (Roche), and 100 U/mL RNaseOUT) and incubated on ice for 10 min. Cells were snap frozen in liquid nitrogen and thawed on ice twice before 10 min centrifugation at max speed. The crude extract (supernatant) was transferred to new tubes and stored at -80°C. Protein concentration was determined by Bradford assay and a small sample of crude extract was run on SDS-PAGE stained with Coomassie Blue to ensure no protein degradation.
In 100 µL reaction, 1.5 µM of PP7-HSPA8 3′ UTR or PP7-LacZ RNA were incubated with 2 µM MBP-PP7 in RNA-IP buffer (20 mM Tris pH 7.2, 200 mM NaCl, 1 mM EDTA pH 8.0, 5 mM dithiothreitol, and 0.01% IGEPAL CA-360) for 1 h on ice. Magnetic amylose beads (100 µL) were washed twice with RNA-IP buffer, then rotated with PP7-HSPA8 3′ UTR or PP7-LacZ bound to MBP-PP7 for 1 h at 4°C. The beads were washed twice with RNA-IP buffer then resuspended in 5 mL RNA-IP buffer supplemented with 0.01 mg/mL tRNA (Sigma, #10109541001) and 5–10 mg N2A crude extract for MS or 2 mg of crude extract for western blots. After rotating the beads and N2A crude extract for 2 h at 4°C, the beads were washed five times with RNA-IP buffer, resuspended in 50 µL RNA-IP buffer and 6 µg of TEV protease, and rotated for 3 h at 4°C. The cleaved PP7 proteins bound to the HSPA8 3′ UTR or LacZ RNA and their interactors was collected and the beads were incubated in fresh RNA-IP buffer containing TEV protease overnight. The elutions were pooled, and the proteins were analyzed by MS as previously described (Proteomics RIMUHC-McGill University)135. Proteins with fold change values > 1.5 and P-values < 0.01 compared to in the control sample were considered HSPA8 3′ UTR interactors. Statistics performed using total spectral count and a T-test analysis.