May 26, 2024
Chromatin accessibility during human first-trimester neurodevelopment – Nature

Chromatin accessibility during human first-trimester neurodevelopment – Nature

Sample collection

All experiments in this study followed all relevant guidelines and regulations, including the International Society for Stem Cell Research 2021 guidelines. Human fetal samples were collected from routine termination of pregnancies at the Karolinska University Hospital, Addenbrooke’s Hospital in Cambridge and the Human Developmental Brain Resource following informed consent of the donors. The use of fetal samples collected from abortions was approved by the Swedish Ethical Review Authority and the National Board of Health and Welfare (Etikprövningsmyndigheten; DNR2020-02074). In the UK, approval from the National Research Ethics Committee East of England, Cambridge Central was obtained as well as approval from the North East – Newcastle & North Tyneside 1 Research Ethics Committee (Local Research Ethics Committee, 96/085; DNR2019-04595). The samples were dissected by a trained embryologist into the major developmental regions (telencephalon, diencephalon, mesencephalon and metencephalon) along the anterior–posterior axis. In addition, the cerebellum was separated from the metencephalon and when possible the metencephalon was divided into medulla oblongata and pons. Following the dissection, the samples were transferred to ice-cold Hibernate E medium (ThermoFisher, A1247601) and either shipped overnight at refrigerated temperature to Sweden or processed the same day when collected at the Karolinska University Hospital.

Some important limitations of this study must be considered. First, as these are clinical samples, the timing was variable and based on expert annotation rather than knowledge of the date of conception. Second, owing to damage incurred during collection, not all regions could be collected from every sample and had to be compensated for by collecting more samples. Third, as the samples were derived from several sources the time between collection and dissociation varied.

Statistics and reproducibility

Sample size was dictated by the availability of scarce early developmental human samples, and based on previous experience with similar studies in mice. No power calculations were carried out to determine sample size. The investigators were not blinded, as this was an exploratory study with anonymous untreated samples. The reproducibility of the dataset across specimens was assessed by the contribution of donors to each cluster (Extended Data Fig. 1d). The linkage disequilibrium score regression analysis linking MDD to midbrain GABAergic neurons was validated in a separate GWAS cohort (see below).

Nucleus isolation

Tissue was gently minced using a razor blade and incubated with the papain dissociation system (Worthington) following the manufacturer’s recommendations (including 200 U ml−1 DNAse), at 37 °C for 10 min. The suspension was then triturated using glass pipettes to dissolve any remaining chunks of tissue, before being filtered through a 30-μm filter (CellTrics). The cells were then washed with EBSS, concentrated (200g, 5 min) and counted using a haemocytometer, after which 1 × 106 cells were pelleted (500g, 5 min) in a 2-ml LoBind Eppendorf tube and pelleted. The cell pellets were dissociated for 5 min on ice using 100 μl of dissociation mix (0.001% digitonin, 0.01% Non-idet P40, 1 mM dithiothreitol, 1 U μl−1 RNAse inhibitor, 0.1% Tween-20, 1% BSA, 10 mM Tris-HCl, 10 mM NaCl, 3 mM MgCl2). When only scATAC-seq was carried out, no RNAse inhibitor or dithiothreitol was added to the mix. Dissociation was halted by addition of 1 ml of wash buffer, after which nuclei were pelleted again (500g, 5 min) and resuspended in 1× nuclei buffer (10x Genomics) and recounted.

Single-cell sequencing

Libraries were generated using the 10x Genomics Chromium Controller and Single Cell ATAC or Single Cell Multiome ATAC + Gene Expression kits. Briefly, a targeted number of nuclei (5,000–10,000) was treated with a Tn5 transposase for 60 min at 37 °C to fragment the DNA and insert adapter sequences into open parts of the chromatin. The suspension was then mixed with the provided barcoding PCR mix and a gel-bead emulsion was generated by co-encapsulating the suspension with barcoded beads in the 10x microfluidic chip and PCR with reverse transcription was carried out in a C1000 Touch thermal cycler (Bio-Rad) with one of two programs—ATAC: 12 cycles of (5 min at 72 °C, 30 s at 98 °C, 10 s at 98 °C, 30 s at 59 °C, 1 min at 72 °C) and hold at 15 °C; or multiome: 45 min at 37 °C, 30 min at 25 °C and hold at 4 °C. For multiome samples, quenching agent was added to prevent the PCR with reverse transcription reaction from continuing. Following PCR, the DNA was isolated from the droplets and cleaned up with Cleanup mix and silane Dynabeads. Sample indices and P7 primers (Illumina) were ligated during library construction using the following PCR protocol: 9 or 10 cycles of (45 s at 98 °C, 20 s at 98 °C, 30 s at 67 °C, 20 s at 72 °C) and 1 min at 72 °C before holding at 4 °C. SPRIselect beads were used for size selection of fragments to generate the final library. The fragment size distribution was analysed using the Bioanalyzer high-sensitivity chip to eliminate libraries that did not show the expected nuclear banding pattern. Libraries were then sequenced using the Illumina Nova-seq instrument using the recommended setting for paired-end sequencing, with the scATAC-seq and scATAC-seq (multiome) libraries in separate flow cells as pooling of them is not recommended with a target of 100,000 read pairs per nucleus. The multiome scRNA-seq libraries were pooled with other 10x Genomics scRNA-seq v3.1 libraries.

10x data processing

All samples were demultiplexed and aligned to the human genome GRCh38.p13 Gencode v35 primary sequence assembly using either Cellranger-atac 2.0.0 or Cellranger-arc 2.0.0 for scATAC-seq and single-cell multiome, respectively. The RNA libraries from multiome samples were aligned as described previously1.

Chromograph pipeline

Chromograph is a new analysis pipeline for scATAC-seq data based on the key architecture of Cytograph 2 (ref. 8), which uses loom files as the underlying data format and is available for use in GitHub (https://github.com/linnarsson-lab/chromograph). The results in this paper were generated using commit #9ae1434. In brief, chromograph provides tools to pool and split scATAC-seq data, carry out clustering, carry out balanced peak calling based on cluster partitions, and identify marker peaks and enriched TF motifs, and enables imputation of gene expression from limited multiome data. This dataset was analysed by first carrying out a primary analysis, and then manually splitting it into subsets based on marker genes. These subsets were then reanalysed, and the results were again pooled to generate a more fine-grained dataset than the primary analysis. The pybedtools and pybigwig packages were used to work with bed files and bigwig files, the loompy package was used to work with loom files, numpy was used to work with matrices and numba was used to speed up computations wherever possible. Scikit-learn and statsmodel were used for more complex calculations and pynndescent was used for fast nearest-neighbour computations.

scATAC-seq quality control

TSS enrichment was calculated using pycisTopic51 (TSS window 50 base pairs (bp), flanking window 1,000 bp) as we noticed discernible change in some of the samples after updating Cellranger-arc. Samples with a score below 5 were discarded. For the other samples, nucleus-by-bin matrices were generated at both 5-kb and 20-kb resolution with bins that overlapped with any of the ENCODE blacklist52 being removed. The 5-kb nucleus-by-bin matrix was used for doublet detection using an adapted version of DoubletFinder. In brief, nuclei were co-embedded with 20% artificial doublets to determine a threshold to distinguish doublets from singlets on the basis of their nearest-neighbour network and a doublet score was assigned on the basis of each nucleus’s local neighbourhood. For the multiome samples, the RNA-doublet score was used as it proved slightly more stable. Additionally, the sex of the sample was determined on the basis of the fraction of Y-chromosomal reads (>0.05% for male) as well TSS fraction. Nuclei that were not doublets and had more than 5,000 and fewer than 100,000 fragments, more than 20% TSS fragments and more than 1,000 RNA unique molecular identifiers (UMIs), and at least 10% unspliced RNA UMIs were pooled to generate the main dataset (the final two filters apply only to multiome).

On average 27,599 high-quality fragments per nucleus were identified with a fragments in peaks ratio of 54%. High-quality nuclei were selected on the basis of the number of fragments and the fraction of fragments overlapping TSS as well as UMI count and splice ratio in multiome samples.

Preclustering and consensus peak calling

The feature set used to generate the nucleus-by-peak matrices is dynamically derived from the data through peak calling. To do so, the 20-kb matrices were first joined and binarized, after which the top 20% of autosomal bins were selected with an upper threshold of 60% coverage across the dataset and decomposed using latent semantic indexing (LSI; more detailed description below). A k-nearest-neighbour graph was then constructed, and the data was clustered into broad clusters using Louvain clustering. Fragments from the nuclei belonging to each cluster were then aggregated and randomly split in two to generate two pseudobulk replicates per cluster. The pseudobulk aggregates were then downsampled to 25 million fragments and MACS253 was used to call peaks using the following parameters: callpeak -f BEDPE -g hs –nomodel –shift 100 –ext 200 –qval 5 × 10−2 -B –SPMR. Peaks were then extended to 400 bp using BEDtools and non-overlapping peaks between the pseudo-replicates were discarded. Next the identified peaks for all clusters were pooled and clustered using BEDtools cluster. For each cluster of peaks, the centre point was extracted and extended to 400 bp to generate the consensus peak set. Peaks overlapping with the ENCODE blacklist were removed and the remainder were annotated using HOMER54 on the basis of Gencode v32, after which the nucleus-by-peak matrix was generated.

Latent semantic indexing

Decomposition was carried out in two steps. First the matrix was depth-normalized and infrequent features were upweighted by carrying out a term frequency–inverse document frequency transformation. The resulting non-binary matrix was then used to compute the principal components using an incremental principal component analysis. Initially 40 components were computed, but components that are not distributed significantly differently from their predecessor are discarded along with a depth-correlated component if present. Next the components were batch-corrected using Harmony to mediate chemistry and sample effects55.

Clustering, embedding and aggregation

The nucleus-by-peak matrix was decomposed using an iterative LSI, meaning that the data was decomposed and clustered in two rounds. First the top 20,000 features by total coverage from the autosomal chromosomes were used to carry out preclustering, after which 20,000 autosomal features were selected again on the basis of the variance of their precluster-level enrichment for a second LSI. Batch effects were again corrected for using Harmony. The second LSI is then used to generate nearest-neighbour graphs and carry out Louvain clustering. A t-SNE map was then generated using an adapted version of ‘the art of using t-SNE’56 that better preserves global structure than native t-SNE. Additionally, a uniform manifold approximation and projection was generated using UMAP-learn57 with default settings. For both methods, Euclidean distances were used as a metric. Next all clusters were aggregated and a normalized counts per million layer was added.

The enrichment of individual peaks was calculated as a Pearson residual58. In brief, fragments were modelled as a negative binomial distribution for which the expected accessibility is the product of the total number of fragments per cluster (c) and the fraction of fragments per peak (g). The residuals can then be calculated as the difference between the observed (Χ) and expected ((widehat{mu })) accessibility corrected by the negative binomial variation (dispersion parameter fixed at 100 for all analysis in this paper). For each cluster, the top 2,000 peaks by Pearson residual were marked as marker peaks. The 20,000 peaks with most variance between Pearson residuals were used to calculate cluster similarities and to generate the cluster dendrogram.

$${Z}_{cg}=frac{{X}_{cg}-{hat{mu }}_{cg}}{sqrt{{hat{mu }}_{cg}-frac{{hat{mu }}_{cg}^{2}}{theta }}}theta =100$$

Gene expression imputation and marker selection

Thirty-one percent of nuclei in the dataset were processed using the Single Cell Multiome ATAC + Gene Expression kit. This allows for the imputation of gene expression measurements in the ATAC-alone samples. To predict gene expression in the scATAC-seq nuclei, first all multiome nuclei were scaled to 5,000 UMIs and an ‘anchor’ net was generated consisting of a directed graph of each scATAC-seq nucleus and their 10 nearest multiome neighbours. Next the weights were scaled to sum to 1 for each nucleus and the nearest-neighbour matrix was multiplied by the gene expression profiles of the multiome nuclei to generate predicted gene expression profiles for each scATAC-seq nucleus.

Trinarization scores and gene enrichment were calculated as defined previously2 with marker genes being selected on the basis of their enrichment. The trinarization scores were then used for auto-annotation using a set of punch cards specific to early human development1,4.

Subset analysis and pooling

The dataset was split on the basis of cluster-level marker expression into the following partitions: fibroblast (DCN and COL1A-expressing), immune (PTPRC-expressing), vascular (TAGLN-expressing or CLDN5 and FLT1-expressing), oligodendrocyte progenitor cell (PDGFRA and OLIG1-expressing), radial glia or glioblast (HES1-expressing or BCAN and TNC-expressing) and neuronal (expressing any of INANHLH1, GAD2SLC17A6 or SLC6A5) lineages. The subsets were reanalysed using the same pipeline described above. Clusters that contained fewer than 10 multiome nuclei or for which less than 1% of the total cluster size was multiome nuclei were excluded as well as clear clusters of doublets. The neuronal lineage partition was split for a second round into GABAergic (GAD2), glutamatergic (SLC17A6, SLC17A7 or SLC17A8) and peptidergic lineages. All partitions were then pooled again and new summary statistics and embeddings were generated.

Motif enrichment

For every cluster, the 2,000 selected marker peaks were used as input to HOMER findMotifsGenome54 using GC-matched genomic sequences as background. The Hocomoco v11 Full collection was chosen as the TF-binding motifs to be tested. The naming convention was manually altered to reflect genes names in the gene expression analysis. This allowed the filtering of false positives by exclusion of nucleus–motif combinations for which the corresponding TF was unlikely to be expressed (trinarization score < 0.5). Additionally, all TFs were assigned to a family on the basis of their arche-motif21.

Gene accessibility and cCREs

Gene-accessibility scores were computed using an adapted version of the cicero workflow59 using the python SKGGM package. First, the distance parameter was estimated by optimizing the calculation of the regularized covariance matrices for 100 random 500-kb regions. Next the distance-adjusted covariance for each accessible region with each TSS site was calculated in 500-kb bins with a 250-kb overlap. Most pairs are sampled twice and pairs with inconsistent covariances are discarded (about 5%). The co-accessibility cutoff was set empirically by testing the number of subnetworks over varying cutoff thresholds. Gene activity scores were then calculated by multiplying the peak-by-nucleus and region-to-TSS covariance matrices, normalizing against size factors derived from a linear regression model and pooling across the 25 nearest neighbours. Similarly to the region–TSS covariance matrix, cCREs were identified by calculating the region–gene expression covariance.

Identification of total accessible regions by sample

To identify general trends in opening and closing of chromatin, all fragments from individual cell classes and biological samples were pooled together and MACS2 was used to call peaks per class per sample. A one-sided Fisher exact test (using the fisher python package) with Benjamini–Hochberg correction was used to identify differential regions. A generalized linear model was used to estimate the influence of age on the number of accessible regions.

VISTA enhancer overlap

CNS enhancers (from the VISTA database19) were downloaded and lifted over to GRCh38 using UCSC liftOver, excluding any that could not be confidently lifted over, resulting in 620 enhancers, of which 596 overlapped with our peak set. The enhancers that were specific to the forebrain, midbrain and hindbrain according to the original authors were isolated (total of 159, 75 and 78, respectively), the corresponding peaks in the dataset were identified and the brain region with the highest accessibility was identified, after which the Jaccard similarity was calculated.

PycisTopic modelling

The full dataset was downsampled to a maximum of 10,000 nuclei per cluster to reduce computational burden and prevent over-representation. The number of topics was varied from 25 to 500 at intervals of 25, running for 50 iterations with an α of 50 divided by the number of topics and a β of 0.1. The most stable model (175 topics) was selected on the basis of topic coherence and log-likelihood in the last iteration. The region-topic scores were normalized so that they summed to 1 for every nucleus and a t-SNE map was generated for the regions and binarized topic lists were generated by assigning each region to the topic that it scored the highest on. Next each topic was used as input for HOMER with the Hocomoco TFs and the results were reduced to the highest-scoring representative of each arche-motif group. The binarized topics were also used as input for Genomic Regions Enrichment for Annotation Tool analysis60 to identify Gene Ontology terms describing each topic. For some selected terms, the associated regions (in the topic) were used to calculate an enrichment score using the signature_enrichment function of pycisTopic51.

Enhancer CNN

Nuclei from all clusters annotated as Purkinje, midbrain GABA, cerebellum granular neuroprogenitor, hindbrain glutamatergic or telencephalic glutamatergic were grouped into five superclusters and enrichment between the clusters was recalculated and peaks were included for learning only if the log-fold change with the second highest accessibility was more than 1. One-hot-encoded sequences (401 bp) were used as input to a CNN trained as a classification model using pyTorch. The network consists of 4 convolutional layers of 256, 60, 60 and 120 nodes and kernel sizes 7, 3, 5 and 3, respectively, and each layer was followed by batch normalization, RELU activation and maximum pooling. There were then 2 dense layers of 256 nodes with batch normalization, RELU activation and a dropout rate of 0.4. A softmax normalization was applied to the final output layer and cross-entropy loss was used as the loss function with label smoothing set to 0.1. The model was trained using an Adam optimizer with a learning rate of 0.01. The model was trained for 26 epochs.

Contribution scores for each sequence were calculated using DeepLiftShap’s (deepExplainer61) attribute function using the mean of the input sequence shuffled 100 times as background. The hypothetical score was calculated for each possible nucleotide in the sequence by multiplying the contribution by the background-corrected input62. TF-MoDisCo62 was then applied to all of the sequences enriched in a cluster with a flanking size of 5 bp, a sliding window of 15 bp and a minimum cluster size of 30 seqlets.

Pseudotime, generalized additive models and ChromVAR

For analysis of the Purkinje lineage, all clusters labelled ‘Purkinje’ and the PTF1A-expressing cluster of ventricular zone progenitors were isolated and a new t-SNE map was generated. pySlingshot was then used to calculate the pseudotime. pyGAM was used to fit gene and cCRE trends to the Purkinje neuron lineage with gene expression being modelled using a Poisson generalized additive model and cCRE accessibility using a linear generalized additive model. ChromVAR was applied using the JASPAR human PWM (human_pwms_v2) to compute motif variability.

Supervised inference and stochastic simulation of Purkinje gene regulatory network

We used DELAY63 (https://github.com/calebclayreagor/DELAY) to infer the Purkinje gene regulatory network from gene-accessibility dynamics in pseudotime and then carried out stochastic simulations to verify the putative network’s gene-expression dynamics. First, we retrained DELAY on a large scATAC-seq dataset of plasma B cell differentiation data64 with ground-truth data from chromatin immunoprecipitation with sequencing65 to prepare the neural network to infer the Purkinje gene regulatory network from tens of thousands of single nuclei. Then, we fine-tuned DELAY on the Purkinje developmental trajectory using ground-truth targets of a cerebellar ataxia-related gene, ataxin 7 (ref. 66). For the final gene regulatory network inference, we used the expression-linked, log-normalized gene-linked peak counts from all TFs that were differentially expressed in at least 1% of Purkinje nuclei across pseudotime (Supplementary Tables 6 and 7; TradeSeq; Wald test; two-sided). We then used BoolODE67 to simulate the expression of each gene in the network given its top eight most likely regulators.

GWAS enrichment

Accessible region locations were lifted over to GRCh37. Features were binarized on the cluster level with a Pearson residual threshold of 10. Cluster heritability was calculated using linkage disequilibrium score regression38. As a background, we used the merger of our feature set with the features from development4 and adulthood39. Only SNPs from hapmap3 were included to reduce imputation errors. In total we tested 325 phenotypes from the UK Biobank26 and 11 psychiatric phenotypes27,28,29,30,31,32,33,34,35,36,37. All used UK Biobank phenotypes had non-zero heritability estimates (z score > 4). Results for UK Biobank phenotype enrichments were corrected for the number of cell types using FDR or Benjamini–Hochberg procedures. For the psychiatric enrichments, FDR and Bonferroni corrections were applied for the number of cell types and tests (α = 3.37 × 10−5, 135 × 11 tests).

Two different MAGMA68 tests were conducted with default settings. First, the cCREs linked to genes were annotated to genes in a custom MAGMA annotation file. A MAGMA gene analysis was used to assess which genes were affected in MDD. Next, MAGMA gene analyses were conducted for ADHD, anorexia, autism spectrum disorder, MDD and schizophrenia, on a custom annotation file in which individual accessible regions were treated like individual genes to identify specific deregulated elements. Accessible regions passing Benjamini–Hochberg correction were then used as input for HOMER with the full vertebrate motif reference.

Reporting summary

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

Source link