Cerebral organoids display dynamic clonal growth and tunable tissue replenishment

Cell culture

Feeder-free hESCs line WA09 (H9) was obtained from WiCell, verified to display a normal karyotype and regularly checked for contamination and mycoplasma. All hESCs were cultured on hESCs-qualified Matrigel (Corning, cat. no. 354277) coated plates with Essential 8 Medium (Thermo Fisher Scientific, A1517001). All stem cells were maintained in a 5% CO2 incubator at 37 °C. Cells were split using DPBS (Dulbecco’s phosphate-buffered saline) −/− (Gibco, 14190-250) to wash once, followed by 5 min incubation at 37 °C, washed off and plated in Essential 8 Medium supplemented with RevitaCell Supplement (Thermo Fisher Scientific, A2644501).

Cloning, molecular biology and generating transgenic and KO hESCs lines

For cell lines expressing fluorophore combination transgenes carrying a CAG-TagBFP-P2A-TagBFP, CAG-TagBFP-P2A-EGFP, CAG-TagBFP-P2A-dTomato, CAG-EGFP-P2A-EGFP, CAG-EGFP-P2A-dtomato or CAG-dTomato-P2A-dTomato cassette were inserted into the AAVS1 locus in H9 cells as previously described41. The modified AAVS1 targeting vector AAVS-SA-2A-puro was a kind gift from R. Jaenisch (Whitehead Institute for Biomedical Research). Nucleofection was carried out as previously described16 using the Amaxa nucleofector (Lonza) with the Stem Cell Kit 1. Cells were grown for 7 days, sorted for fluorophore expression and plated as single cells. Colonies were picked and genotyped to confirm transgene presence. For KO cell line generation, the pSpCas9(BB)-2A-GFP (PX458, Addgene: 48138) plasmid, modified to express eCas9 instead of WT-Cas9 and dTomato instead of GFP, was used carrying a gRNA targeting ASPM, TP53 or PAX6. H9-CAG-EGFP-P2A-EGFP cells were nucleofected as described above, sorted by flow cytometry for dTomato expression after 3 days and plated at low density on a 10 cm dish. Single-cell colonies were picked and verified to carry frame-shift mutations.

Cerebral organoid generation

Cerebral organoids were generated in our lab as previously developed19,20. Briefly, hESC cells were grown to 60–80% confluency and single-cell suspensions were obtained using Accutase. Pelleted cells were resuspended in hESC media supplemented with RevitaCell and counted. In total, 24,000 cells and 5–10 poly(lactide-co-glycolide) copolymer (PLGA) fibre microfilaments were seeded to form embryoid bodies in 96-well ultra-low-attachment U-bottom plate (Sigma, CLS7007) in 150 μl E8 supplemented with RevitaCell. On day 3, media was changed to E8, and from day 6 on organoids were grown as previously described4,6. In experiments that included puromycin treatment, media was supplemented with 0.3 μg/ml Puromycin from day 11 on (Sigma-Aldrich, P8833). For some experiments, organoids were incubated with 100 µM BrdU (Sigma, B5002) for 1 h followed by washing 24 h prior to analysis.

Differentiation of neurons in 2D

At day 0, EBs were formed as described above directly in N2B27 differentiation media supplemented with 10 µM SB 431542 (Tocris, 1614), 0.25 µM LDN193189-hydrochlorid (Sigma-Aldrich, SML0559) and 10 µM RevitaCell in 96-well ultra-low-attachment U-bottom plates. N2B27 differentiation media was 1:1 Dulbecco’s Modified Eagle Medium:Nutrient Mixture F-12 (DMEM/F12, Gibco, 12330-057):Neurobasal media (Gibco, 21103049) supplemented with N-2 Supplement (100×, Thermo Fisher Scientific, 7502048), B-27 Supplement (50×, Thermo Fisher Scientific, 17504044), NEAA (Sigma-Aldrich, M7145) and GlutaMAX (Thermo Fisher Scientific, 35050-038). On days 2, 4 and 6 media was changed with N2B27 media supplemented with 10 µM SB, 0.25 µM LDN. On day 7 EBs were plated on 96-well plates coated with growth factor reduced Matrigel (Corning, 354230) in N2B27 media. Media was changed every second day with N2B27 media. On day 21 plated EBs were dissociated using Accutase, and pelleted cells were resuspended in N2B27 media supplemented with 10 ng ml–1 human recombinant brain-derived neurotrophic factor (BDNF, Stemcell Technologies, 78005), 10 ng ml–1 human recombinant glial cell line-derived neurotrophic factor (GDNF, Stemcell Technologies, 78058) and 10 µM DAPT (Stemcell Technologies, 72082) on poly-l-ornithine (Sigma-Aldrich, P3655) and laminin (Thermo Fisher Scientific, 23017015) coated 24-well plates. Plates were coated with poly-l-ornithine for 1 h at 37 °C, washed three times with DPBS and recoated with 10 µg ml–1 laminin for 2 h. Media was changed every second day with N2B27 media supplemented with 10 ng ml–1 BDNF, 10 ng ml–1 GDNF and 10 µM DAPT (Sigma-Aldrich, D5942). From day 31 on DAPT is left out.

In utero injection

For this study, we used 10–12-week-old timed pregnant C57BL/6J mice (Jackson Laboratory). Mice were kept at 22 °C +/− l °C, 55% +/− 5 % humidity in a 14 h light/10 h dark cycle under the care of the Institute of Molecular Biotechnology (IMBA) animal care facility. For in utero injections mice were anaesthetized, uterine horns were exposed, and concentrated, Eco-coated retrovirus was injected into the ventricle of E9.5, E10.5 or E13.5 embryos followed by suturing. At E17.5 left and right hemispheres of the cortex and midbrain were harvested. Injections at days E9.5 and E10.5 were guided by ultrasound visualization (Vevo 770, scanhead RMV711, Visualsonics). Embryos were not tested for sex. All procedures were performed in accordance with protocols approved by the Austrian Federal Ministry of Education, Science and Research under the animal experiment licence BMWF.66.015/0023.WF/3b/2017. 3R principles (replacement, reduction and refinement) were followed and monitored by the IMBA Ethics and Biosafety Department.

Virus construction and viral library preparation

Two retroviral libraries based on pRSF retrovirus expressing TagBFP and containing one of two semi-random barcode libraries were used for lineage tracing. To generate the random barcode libraries, library oligos (Supplementary Table 1) were amplified until they reached logarithmic phase (6–8 cycles, monitored in qPCR machine, stopped in the extension phase). The purified PCR product was cloned into a modified pRSF retroviral vector using XhoI and EcoRI restriction enzymes. Purified ligation reactions were transformed into MegaX DH10 T1R Electrocomp Cells (Thermo Fisher Scientific, C640003) according to the manufacturer’s instructions and plated onto lysogeny broth (LB) media plates containing ampicillin. Using dilution series libraries were determined to contain at least 20 × 106 colonies. Overnight colonies were scraped and recovered for 1 h in LB media containing ampicillin. Plasmid DNA was extracted using a QIAGEN EndoFree Plasmid Maxi Kit (12362).

Virus production

For WT lineage tracing experiments, plasmid libraries were mixed prior to making retrovirus. As both libraries acted highly similarly in WT they were used separately to distinguish individual genotypes in chimeric organoid experiments. Retrovirus was produced in HEK293 grown in DMEM/10% fetal bovine serum/2 mM l-glutamine/100 units ml–1 penicillin/0.1 mg ml–1 streptomycin and coated using vesicular stomatitis virus G (human cells) or Eco-R (mouse). Supernatant was collected for 72 h every 12–24 h, pooled, pelleted at 28,000g and resuspended in stem cell medium containing 4 μg ml–1 polybrene (Sigma-Aldrich, H9268) and 100 μg ml–1 Synperonic F108 (Sigma-Aldrich, 07579) (hESCs) or Hanks’ balanced salt solution (mouse). Human cells were split 1:2.5 1 h prior to infection using Accutase. Infections were performed for 3 h, followed by two washes using DMEM/F12 and addition of fresh Essential 8 medium supplemented with RevitaCell.

Lineage tracing with genomic barcoding

Virus was produced and hESCs and mice infected. One to two days after infection single stem cell suspensions were prepared, sorted by flow cytometry for TagBFP (4–7% TagBFP-positive, 1.3–1.8 × 106 cells total sorted, actual infection rates were determined to be up to 19% on not previously sorted cells three days after infection) and plated in E8 supplemented with RevitaCell to recover for 1–2 days before organoid setup. Minimal times between viral library infection, sorting and organoid setup were kept to prevent expansive clone growth. Organoids, stem cells and mouse brains (E17.5) were harvested at days 0–42. Samples were lysed in a single reaction vessel in 400 µl gDNA extraction buffer (10 mM Tris pH 8, 150 mM NaCl, 10 mM EDTA, 0.1 % sodium dodecyl sulfate (SDS) and 0.2 mg ml–1 Proteinase K) incubated at 55 °C O/N followed by 85 °C for 1 h. RNase A (1 µl; Thermo Fisher Scientific, EN0531) and RNase T1 (0.2 µl; Thermo Fisher Scientific, EN0541) were added, incubated at 37 °C for 2 h followed by phenol/chloroform extraction and EtOH precipitation. After 12 freeze/thaw cycles viral barcode inserts were amplified using lineage tracing genomic DNA primers with sequencing adaptors. Libraries were sequenced on Illumina HiSeq 2500 and Novaseq S1 lanes. At least three replicates per condition and time point were analysed. Out of 476 samples sequenced in total, 20 were excluded from further analysis due to inefficient PCR amplification. In spite of exclusion of those samples, at least two measurements per experimental condition were used for analysis in all cases22.

Dissociating organoids for flow cytometry analysis

For flow cytometry analysis organoids were dissociated in a 10:1 mixture of Accutase (Sigma-Aldrich, A6954) and 10× Trypsin (Gibco, 15400) shaking at 37 °C and filtered through a 35 µm strainer. Aliquots were counted on an Invitrogen Countess II cell counter. Antibody staining was performed in DMEM/F12 + 4% BSA using the following antibodies: anti-NCAM (BD Biosciences, 564058), anti-CXCR4 (BD Biosciences, 560936) and anti-TRA-1-60 (BD Biosciences, 563188).

RNA extraction and qPCR

RNA samples and qPCR was performed as described previously25. In short, RNA samples were processed using the Qiagen RNeasy kit (Qiagen, 74004). cDNA was generated using Superscript (Invitrogen, 18080044) and qPCR was performed using Promegas qPCR master Mix (Promega, A6002). Primers used were published previously25.

Western blotting

For TP53 induction, Nutlin-3a (10 μM) was added to organoid culture media for 24 h prior to lysis. Organoids were lysed with radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, 89900) with complete protease inhibitor (Roche, 05056489001). Protein was quantified by bicinchoninic acid assay (Thermo Fisher Scientific, 232250) and 25 μg of sample was denatured in reducing Laemmli buffer (Thermo Fisher Scientific, J61337) before protein separation by SDS–PAGE on 4–12% Bis-Tris gels in 3-(N-morpholino)propanesulfonic acid (MOPS) SDS buffer (Invitrogen, NP0323BOX, NP0001). Proteins were transferred to nitrocellulose membranes and incubated in 4% skimmed milk with primary antibody including anti-LaminB1 (1:2,000, GeneTex, 103292), and either anti-PAX6 (1:1,000, Biolegend, 901301) or anti-TP53 (1:1,000, AbCam, ab32389). Species-specific secondary antibodies (both 1:10,000, Li-cor, 68070, 32211) were used to detect bands by fluorescence (BioRad ChemiDoc MP).

Cryosectioning and immunofluorescence

For histological staining, tissues were fixed overnight in 4% paraformaldehyde. For cryosectioning fixed organoids were embedded in OCT (Sakura, 4583), cut at 18–20 μm thickness and processed for immunofluorescence using standard methods. The following primary antibodies were used: anti-SOX2 (Abcam, ab97959, 1:500), anti-NeuN (Millipore, MAB377, 1:500), anti-DCX (Santa Cruz, sc8066, 1:1,000), anti-MAP2 (Abcam, ab5392, 1:1,000), anti-Nestin (BD, 611658, 1:1,000), anti bIII-tubulin (Abcam, ab18207, 1:1,000), anti-BrdU (Thermo Fisher Scientific, B35128, 1:500), anti-phospho-vimentin(Ser82) (MBL, D095-3S, 1:250), anti-TTR (AbD Serotec, ahp1837, 1:500), anti-aPKC (SantaCruz, sc-17781, 1:100), anti-nestin (Abcam, ab105389, 1:1,000), anti-β-catenin (Sigma, C2206, 1:250), anti-N-cadherin (BD, 610920, 1:500), anti-ASPM (Bethyl Laboratories, IHC-00058, 1:100), anti-TBR2 (Abcam, ab23345, 1:250) and anti-TBR1 (Abcam, ab31940, 1:500). Secondary antibodies raised in donkey or goat were purchased from Thermo Fisher Scientific: donkey anti-mouse immunoglobulin G (IgG) (H+L), Alexa Fluor 488, A-21202, 1:1,000; donkey anti-rabbit IgG (H+L), Alexa Fluor 488, A-21206, 1:1,000; donkey anti-goat IgG (H+L), Alexa Fluor 488, A-11055, 1:1,000; donkey anti-sheep IgG (H+L), Alexa Fluor 488, A-11015, 1:1,000; goat anti-rabbit IgG (H+L), Alexa Fluor 488, A-11008, 1:1,000; goat anti-mouse IgG (H+L), Alexa Fluor 488, A-11001, 1:1,000; goat anti-mouse IgG1, Alexa Fluor 488, A-21121, 1:1,000; goat anti-rabbit IgG (H+L), Alexa Fluor 568, A-11011, 1:1,000; goat anti-mouse IgG (H+L), Alexa Fluor 568, A-11004, 1:1,000; donkey anti-goat IgG (H+L), Alexa Fluor 568, A-11057, 1:1,000; donkey anti-mouse IgG (H+L), Alexa Fluor 568, A-11037, 1:1,000; goat anti-mouse IgG2b, Alexa Fluor 568, A-21144, 1:1,000; donkey anti-chicken IgY (H+L), Alexa Fluor 568, A-78950, 1:1,000; donkey anti-rabbit IgG (H+L), Alexa Fluor 647, 31573, 1:1,000; donkey anti-goat IgG (H+L), Alexa Fluor 647, 21447, 1:1,000; goat anti-rabbit IgG (H+L), Alexa Fluor 647, 21245, 1:1,000; and goat anti-rabbit IgG (H+L), Alexa Fluor 647, 21244, 1:1,000. If possible, all secondary antibodies were of highly cross-adsorbed quality.

Imaging and microscopy

Imaging was performed on Zeiss (LSM 780, 800, 880, Z1) or Olympus (IX83) microscopes. Channels were collected individually with excitation being delivered by lasers of 405, 488, 561 and 633 nm wavelength. Alternatively, imaging was performed on Panoramic250 slidescanners from 3DHistech. Images were processed in the respective companies’ image suites and FIJI. All equipment was kept in immaculate condition by the IMP/IMBA biooptics facility.

Transduction of cerebral organoids for live imaging

Cerebral organoids (week 6) were embedded in 3% low melting point agarose dissolved in DMEM/F-12. Organoids were sliced with a Leica VT1200S vibratome in ice-cold DMEM/F-12 into sections of 250 µm. Sections were transferred to a 24-well plate, transduced with a GFP-encoding retrovirus (MSCV-IRES-GFP: Addgene 20672) or adenovirus (Vector Biolabs, 1060) and incubated for 2 h at 37 °C. After four washes with DMEM/F-12, the slices were transferred to Millicell cell culture inserts (Merck) and placed in a six-well plate containing cortical culture medium (DMEM/F-12 containing B27, N2, 10 ng ml–1 fibroblast growth factor, 10 ng ml–1 epidermal growth factor, 5% fetal bovine serum and 5% normal horse serum). Slices were cultured for 2 days before live imaging.

Live imaging of cerebral organoids

Prior to live imaging, culture inserts containing the organoid slices were transferred to a six-well glass bottom plate (Cellvis) and fresh cortical culture medium was added. GFP-positive cells were live imaged for 48 hours on a spinning disk wide microscope equipped with a Yokogawa CSU-W1 scanner unit. The microscope was equipped with a high working distance (WD 6.9–8.2, 414 mm) 20× Plan Fluor ELWD NA 0.45 dry objective (Nikon), and a Prime95B SCMOS camera. Z-stacks of 80–100 µm range were taken with a step size of 4–5 µm at intervals of 15 minutes. Temperature and CO2 levels were controlled with a stage top incubator (Tokai Hit). Raw images were processed with NIS-Elements and FIJI ImageJ.

Immunostaining after live imaging

Cerebral organoid slices were fixed in 4% PFA for 20 minutes. Slices were blocked and permeabilized in 5% normal donkey serum 0.25% Triton X-100 for 1 hour at room temperature. Primary and secondary antibody mix was prepared in 5% normal donkey serum 0.1% Triton X-100 and incubated overnight at 4 °C. Samples were mounted in VECTASHIELD HardSet (Vector Laboratories) and mosaics (3 × 3 tiles) were acquired with CFI Apo LWD Lambda S ×40 objective (NA 1.15; WD 0.61–0.59, Nikon). Primary antibodies used included: sheep anti-EOMES (1:200, R&D Systems AF6166), chicken anti-GFP (1:500, Abcam Ab13970), mouse anti-Sox2 (1:500, Abcam Ab79351) and rabbit anti-NeuroD2 (1:500, Abcam Ab104430).

Live-fixed correlative microscopy

Correlative microscopy was performed as described previously28. In brief, live and fixed images were segmented, paired and aligned using an ImageJ and MATLAB-based macro. Once positional information was retrieved and transferred to the fixed images, high-resolution mosaics (3 × 3 tiles) were acquired and cell fate was assessed using canonical cell fate markers.

Barcoded scRNA-seq

A plasmid library was cloned as described above to insert a 20 bp semi-random barcode into the 3′ UTR (untranslated region) of a Pol II transcript encoding GFP (sequences in Supplementary Table 1). The purified PCR product containing the barcode was cloned into the backbone using EcoRI and NheI restriction sites. Infected hESCs were as described above. After 48 hours, GFP-barcoded cells were mixed with BFP-expressing cells, which had been transduced with an equivalent, non-barcoded retrovirus. Cells were mixed at a ratio of 5:95 (GFP:BFP) and were seeded to generate organoids.

Organoids were dissociated at day 18 or 42 as before, filtered through a 35 μm strainer followed by dilution with 0.1% BSA in DPBS –/– and stained with Draq7 viability dye (Biostatus; DR70250, 0.3 mM). Live, single, GFP-positive cells were sorted with a BD FACSAria III (70 nm nozzle) and collected in a 96-well PCR plate pre-loaded with 10 μl of 0.5% BSA in DPBS –/–. Single organoids were sorted to completion into a single well and the entire resulting cell suspensions were subjected to scRNA-seq analysis using the Chromium Single Cell 3′ Reagent Kit (v.3.1) following the 10x Genomics user guide with the following modification. During cDNA amplification (2.2), the PCR reaction was paused at 72 degrees after six cycles. An aliquot (75 μl) of sample is removed for targeted amplification and replaced with 37.5 μl of Amp Mix, 11.25 μl cDNA Primers and 26.25 μl H2O before continuing for a total of 13 cycles. Targeted amplification to enhance barcode recovery was performed as described previously38. Briefly, sixth cycle cDNA from 10x libraries were PCR amplified in a hemi-nested manner using limited PCR cycles with AmpliTaq Gold 360 (Thermo Fisher Scientific, 4398876). Sequencing of 10x gene expression libraries and targeted amplification libraries was performed on an Illumina NovaSeq S4 lane.

scRNA-seq analysis

Per sample, sequenced reads were aligned to the GRCh38 2020-A human reference genome with Cell Ranger version 7.01 (10x Genomics) using default parameters. Resulting cell-by-gene, unique molecular identifier (UMI) count matrices were analysed in R using Seurat version 4.3.0.1. We filtered for high-quality cells based on doublet detection performed with scrublet (version 0.2.3), number of uniquely detected genes (‘nFeature’), number of UMIs (‘nCount’), percentage of mitochondrial reads and percentage of ribosomal reads. The latter four thresholds were set for each sample separately and are listed in Supplementary Table 2. Subsequently, the remaining high-quality cells were merged for day 18 and day 42 samples. The following analysis was performed for both timepoints: counts were log-normalized cell-wise (‘LogNormalize’), 2,000 highly variable genes were identified (‘FindVariableFeatures’) and scaled (‘ScaleData’). Next, principal component analysis was performed (‘RunPCA’) on a custom gene list used previously25 and based on the first 50 components a UMAP embedding was computed (‘RunUMAP’). To cluster the data, the shared nearest neighbour graph was calculated (‘FindNeighbors’, also based on the first 50 principal components) and used as input for the Louvain clustering algorithm (‘FindClusters’, resolution 1.2 for day 18 and 0.7 for 42). Clusters were annotated based on expression of known marker genes for radial glia, intermediate progenitors and neurons.

Bioinformatic analyses

The following sequencing information was extracted from the raw reads: the 50-base-long viral barcode was extracted from read 1; the sample barcode was extracted from index 1 and 2. Sample barcodes were error-corrected by comparison to known input sequences, while base errors in viral barcodes were corrected by a clustering algorithm42. Lineage barcodes that were found in a different sample with a 20× or higher read count were removed from the sample with the lower read count. Lineage barcodes with a read count lower than a sample-specific threshold are removed. Sample-specific thresholds are chosen as suggested previously43. Samples for which lineage barcodes overlapped more often with lineage barcodes of other samples than expected or with low lineage or read count were removed as outliers.

Optimal transport

To compare the lineage size distributions of two samples and we used an optimal transport approach. If two samples had a different read threshold, the higher read threshold was applied to both samples. Both samples were normalized to reads per million. If the number of lineages differed between the two samples, each lineage in sample xi was counted as lcm(N, M)/N lineages and each lineage in sample yi as lcm(N, M)/M lineages. We then find a transport map T(x) that maps each lineage xi to a lineage T(xi) = yi so that the sum of fold changes of the lineage sizes \(\sum \,\log_2(}}_}})-\,\log_2(}(}}_}}))\) is minimal. The solution to this optimal transport problem is given by mapping rankwise, so that the lineage with rank i in sample x(i) is mapped to the lineage with rank i in sample T(x(i)) = y(i). For the fold-change curves, the top 1% lineages were removed due to high graph variability, after which the graph is resampled and smoothed with LOWESS regression and averaged over all replicates.

Modelling

A lineage is modelled as its number of S cells (s), A cells (a) and N cells (n). Three events can modify the cell counts: symmetric division (+1 S cell), transition (–1 S cell, +1 A cell) and asymmetric division (+k N cells) with corresponding rates rs, ra and rn. Each lineage is independently simulated stepwise. In each step the number of events is approximated by the binomial distribution with mean rx where r is the rate of the event and x is the number of originating cells (that is, S cells for symmetric division, S cells for transition and A cells for asymmetric division). The time step is chosen as 0.01/max(rs, ra, rn) so that the probability of the same cell dividing twice in one time step (which the binomial distribution does not capture) is smaller than 1%. An organoid is simulated as 10,000 independent lineages.

Statistics and reproducibility

No statistical method was used to predetermine sample size but our sample sizes are similar to those reported in previous publications16,19,25. No data were excluded from analyses with the exception of low-quality sequencing libraries as indicated in the corresponding Methods section. Experiments were not randomized, and investigators were not blinded to allocation during experiments and outcome assessment. Statistical testing in this study (Kolmogorov–Smirnov) does not require normal distribution of the data; therefore it was not formally tested. At least two independent biological experiments were performed including multiple replicates for most experiments with comparable results. Exceptions include some whole organoid lineage tracing experiments, which were conducted only once with three individual organoids per timepoint and conditions and four mice per timepoint. Barcoded scRNA-seq experiments were performed from one batch with one organoid per 10x library in organoid duplicates per two timepoints. Micrographs show representative images from at least two experiments including multiple organoids.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

留言 (0)

沒有登入
gif