Arid1a cKO mice exhibit disorganized dentate gyrus
To explore the role of Arid1a in the central nervous system, we first examined its expression patterns in neural stem/progenitor cells (NSPCs), neurons and astrocytes. We performed ARID1A immunostaining of neural lineage cells by using nestin-GFP and Thy1-GFP transgenic mice in which NSPCs or neurons were labeled with the green fluorescent protein (GFP). The results showed that ARID1A localized mainly in the Nestin-positive NSPCs and Thy1-positive neurons (Supplemental Fig. S1A). We then co-stained ARID1A with astrocyte specific maker GFAP and found that ARID1A was also expressed in the astrocytes (Supplemental Fig. S1A). Next, we examined the expression of Arid1a at different embryonic and postnatal stages and found that Arid1a was highly expressed at the prenatal stage, whereas both mRNA and protein levels of Arid1a were gradually decreased after birth (from P0 to P21) (Supplemental Fig. S1B-D). Therefore, these results suggest that Arid1a involve in NSPCs and neural development.
To determine the function of Arid1a in the developing brain, we generated Arid1aflox/flox; Emx1-cre mice (hereafter referred to as Arid1a cKO) by crossing Arid1aflox/flox mice with Emx1-Cre transgenic mice to delete Arid1a specifically within the cortex and hippocampus (Supplemental Fig. 1E). Immunofluorescence staining (Fig. 1A) and western blotting (Fig. 1B, C) analyses from forebrain tissues at embryonic day 16.5 confirmed that Arid1a was successfully deleted. To study the integrity of SWI/SNF after Arid1a deletion, we tested the expression of BRG1, the central ATPase subunit of SWI/SNF. The western blotting results showed that BRG1 significantly down-regulated after Arid1a specific deletion in the forebrain (Fig. 1B, C). Arid1a cKO mice were born at the expected Mendelian ratios and were indistinguishable from their WT littermates at postnatal day 0 (P0). However, most Arid1a cKO mice (> 80%) died at 4–5 weeks after birth (Supplemental Fig. S1F). Histologic analysis of cKO mice revealed striking abnormalities in the hippocampal formation at P21 (Fig. 1D), which lacked the dentate granule cell layer and displayed disorganized dentate hilus and pyramidal layers (Fig. 1D). We then examined the granule cell layer (GCL) in Arid1a cKO mice at prenatal (E16.5 and E18.5) and postnatal stages (P0, P7, P14 and P21). In Nissl-stained sections, the developing pyramidal cell layer in Ammon’s horn of the hippocampus was similar in morphology (such as breadth) between Arid1a cKO mice and their WT littermates (Fig. 1E). By contrast, the DG region in cKO mice was hardly discriminated from E16.5 to E18.5, and displayed a significantly smaller GCL volume at P0 compared with WT mice (Fig. 1E). Moreover, the difference GCL volume between cKO and WT mice further magnified as they grew older (Fig. 1E, F).
Development of DG involves various cell types including NSPCs and granule cells, therefore the cell compartments in the DG of Arid1a cKO mice are highly likely altered as their remarkably reduced size. The number of BLBP+ NSPCs in Arid1a cKO mice was significantly decreased at P0 compared with the WT controls (Supplemental Fig. 2A, B). In addition, the numbers of calretinin + mossy cells and calbindin + granule cells were all significantly reduced in the DG area of cKO mice at P21 (Supplemental Fig. 2C-F). These data suggest that Arid1a is essential for DG development.
Loss of Arid1a damages neuronal cell composition in hippocampus
Distinct lamination appeared in the dentate gyrus at P726. Dendritic spines begin to increase in density during the first week after birth and with a peak in the third week27. We thus performed scRNA-seq (10x Genomics platform) to examine the abnormality in the Arid1a cKO hippocampus at P7 from three mice. After quality control and filtering, 14,894 cells from cKO samples and 15,520 cells from WT samples were used for further analysis. To identify the major cell types in the hippocampus, we used unsupervised clustering and identified 11 major distinct clusters according to the expression of canonical gene markers (Fig. 2A). These cells included progenitor cells (Mki67, Ascl1), astrocytes (Gfap, Aqp4), neurons (Neurod2, Rbfox3), microglia (Cx3cr1, P2ry12), oligodendrocyte precursor cells (OPCs) (Olig2, Pdgfra), Cajal-Retzius cells (Ndnf, Clstn2), ependymal cells (Ccdc153, Dnah11), endothelial cells (Cd93, Arhgap29), blood (Hba-a1, Hbb-bs), mural cells (Col3a1, Arhgap29) (Fig. 2C and Supplemental Fig. 3A). Cells clustered largely by connections between different cell types (Supplemental Fig. 3B).
To reveal the differences in cell compositions between WT and cKO, we analyzed the relative percentage of the 11 major cell types based on scRNA-seq data. The relative percentage of progenitors decreased 10% in cKO mice. Of note, the percentage of neurons decreased 20% in cKO mice, compared with WT mice. The massive decrease of neurons was in accordance with the smaller volume of hippocampus in cKO mice (Fig. 2B, D). Next, to investigate the transcriptomic changes of different cell types in cKO mice, we performed differentially expressed genes (DEGs) analysis (Supplemental Fig. 3C). We found progenitor cells exhibited differential expression of Ccnd1, Ccnd2, Nrn1, and Npy, which were involved in mitotic cell cycle, cell cycle phase transition and cell differentiation (Fig. 2E, F). These data indicate that Arid1a is required for NSPCs proliferation.
Arid1a regulates neuronal dynamic changes in DG
To characterize gene expression changes in neurons between WT and cKO mice, we first identified 2 groups according to known cell type and developmental markers (Fig. 3A). Elavl2, Dkk3, Homer3, and Pcp4 which have been identified as markers of cornu ammonis (CA), while Prox1 was expressed in DG (Supplemental Fig. 4A). To further confirm the lineage relationships and neurons in the DG region are most affected, we used Monocle analysis. Monocle2 identified a bifurcating trajectory with three branches. Progenitor cells (PCs) were the beginning and end of the trajectory at two branches, and neurons in CA or DG were distributed at the other end of the trajectory branch, indicating their neuronal identity (Fig. 3B). Using pseudo-time analysis, we found Elavl2, Pcp4, Prox1 expression were gradually upregulated and Sox2 expression was downregulated along pseudo-temporally ordered paths from progenitor cells to neurons (CA/DG) (Fig. 3C). To further uncover the different regulating modules of gene expression during the development of neurons, we clustered genes using Monocle 2. Three different gene expression modules along with pre-branch(root), cell fate1(state 1–2), cell fate 2(state 3) of neuron development were identified by branched expression analysis modeling (BEAM) for significantly regulated genes. In addition, we defined three distinct development stages based on the three different gene expression modules, including the naïve stage (module 1), intermediate stage (module 2), and mature stage (module 3) (Supplemental Fig. 4B).
To help in determining the biological processes of neurons development, Gene Ontology (GO) term enrichment (top three GO terms) was performed for three different gene expression modules(Supplemental Fig. 4B). Module 1 was mainly comprised of gene sets that were involved in the biological process of the mitotic cell cycle(Mki67, Cdk1, Ccnd2, Hmgb1), DNA packaging (Top2a, Cdk1, Hmgb1), which played a vital part in cell proliferation. The genes enriched in Module 2 were involved in the positive regulation of neuron differentiation (Id4, Hes5, Mmd2), axon ensheathment in central nervous systems (Apoe, Id4, Hes5). In module 3, the enriched genes were upregulated in cell fate 2(state 3), which was involved in synapse maturation (Grbra2, Grin2b, Nrn1, Reln, Neurod2), granule layer formation (Nrxn1, Prox1, Dcx, Nfix,), dentate gyrus development (Prox1, Reln, Neurod6), cognition (Nrxn1, Meis2, Reln, Neurod2, Grin2b). In summary, the common and distinct gene regulation patterns were constructed in neuron development, which contributed to an in-depth understanding of dentate gyrus development processes and underlying regulatory basis.
Genes transiently regulated at the bifurcation point where neuronal lineage separated from PC lineage could have a critical function in promoting the commitment of progenitor cells. Using Monocle2, we found that the expression of Prox1 was significantly decreased in the neuron of DG in Arid1a cKO mice compared with WT mice, while the markers of CA such as Elavl2, Pcp4 were decreased slightly in Arid1a cKO mice compared with WT mice (Fig. 3D). In addition, we found that neurons of DG displayed differential expression of Prox1, Calb2, Rbfox3, Casp3 and Bax, which were associated with granule cells development, neuron differentiation and cell apoptosis (Fig. 3E and Supplemental Fig. 4C). Together, our data therefore indicate a sequential activation of Elavl2, Pcp4, Prox1 during PC commitment and confirmed a strong change in cell differentiation and cell apoptosis in Arid1a cKO mice DG.
Arid1a is required for the proliferation and differentiation of NSPCs
To further confirm the phenotypes of Arid1a deficiency on DG development, we compared the number of dividing cells (Ki67+ cells) in the DG region in Arid1a cKO and WT littermates. The results showed that Ki67+ cells decreased dramatically in Arid1a cKO mice at all developmental stages (Fig. 4A, B). To examine whether Arid1a deficiency alters the proliferation and division of neural stem and progenitor cells (NSPCs) in DG, the mice received a single intra-peritoneal (i.p.) injection of bromodeoxyuridine (BrdU), and brains were collected 2 hours later. The quantification results demonstrated that the number of BrdU+ cells was significantly reduced in SGZ of Arid1a cKO mice from E16.5 to P21 (Fig. 4C, D). These results indicate a pivotal role of ARID1A in the proliferation of NSPCs.
To evaluate whether Arid1a cKO also affects neural differentiation in DG, we first analyzed the expression of DCX in the DG of Arid1a WT and cKO mice. In support of this, we also found that Arid1a cKO mice had fewer DCX+ cells (types 2b and 3 NPCs and immature neurons) in the SGZ (Fig. 3E, F). These results clearly suggest that Arid1a is required for neural differentiation and cell fate determination in vivo. Furthermore, we found that the Arid1a cKO mice brains displayed drastically increased apoptosis in the DG compared with their littermate controls (Fig. 4G, H). Taken together, these data showed that the deletion of Arid1a impaired the proliferation and differentiation of NSPCs in the dentate gyrus and induced cell apoptosis.
To further assess the function of Arid1a in neurogenesis, we isolated hippocampal NSPCs from postnatal Arid1a cKO mice and their WT littermates. To assess the proliferation of NSPCs, we pulse-labeled the cells with BrdU for 2 hours (Fig. 5A). Quantification of BrdU+ Nestin+ labeled cells demonstrated that less cKO NSPCs had incorporated BrdU than that of WT NSPCs (Fig. 5A, B). Moreover, in an in vitro assay, we isolated hippocampal NSPCs from Arid1a cKO or WT pups at postnatal, and cultured them in the neural differentiation medium. We observed a decreased number of Tuj1+ cells that was differentiated from Arid1a cKO NSPCs (Fig. 5C, D). These results indicated that Arid1a played essential role in NSPCs proliferation and cell differentiation in hippocampus in vitro.
Loss of Arid1a results in profiling changes in histone modifications and abnormal gene transcriptions
To investigate the consequences of altered SWI/SNF targeting induced by Arid1a deletion, we characterized histone modifications associated with cis-regulatory elements (H3K4me3, H3K27me3, and H3K27ac) at SWI/SNF binding sites in the hippocampus of wild-type and Arid1a cKO mice at E16.5. As expected, most of these three histone markers are enriched around the TSS of protein coding genes (Fig. 6A). While, among these three histone markers, H3K4me3 and H3K27ac downregulate the enrichments in TSS, H3K27me3 upregulates the enrichment (Fig. 6A). H3K4me3 and H3K27ac modifications enrichment at the TSS regions is important for gene activation. The Venn diagram (Fig. 6B) and Gene Ontology (GO) enrichment (Supplemental Fig. 5A) analysis of 47 downregulated genes with H3K4me3 and H3K27ac modification showed enrichment in the pathways involved in dentate gyrus development, neuron differentiation and synapse maturation.
To identify the underlying molecular mechanism, we examined the effect of Arid1a deletion on Brg1 expression. We found that Brg1 protein levels decreased in the cKO group (Fig. 1B, C). Because components of the BAF complex contribute to the specificity of the BAF complex, these results indicate that Brg1 recruitment may be affected by Arid1a deletion. Based on previously published data, we found that Brg1 also accumulates at the TSS region (Fig. 6A). These data suggest that Arid1a may be required for proper recruitment of Brg1 to maintain proper nucleosome configuration for gene expression in the hippocampus.
To further elucidate gene regulatory mechanisms underlying the regulation of Arid1a on the hippocampus development, we integrate H3K4me3 ChIP-seq, H3K27ac ChIP-seq, and scRNA-seq DEGs in progenitor cells and neurons to enrich the target genes under the epigenetic modulations (Fig. 6B). Go analysis (Biological of Process) show that these 46 overlapping DEGs are involved in the regulation of organ growth, dentate gyrus development, granule cells differentiation (Fig. 6C). Especially Prox128, Tmem10829 have been reported to be involved in the regulation of DG development. During all the examined targets (Fig. 6D), we mainly focus on the homeobox gene Prox1, which is highly expressed in several brain regions (i.e., cortex, DG, thalamus, hypothalamus, cerebellum) during prenatal and postnatal stages of development 30, 31. Published data have shown that Prox1 is expressed through all the stages of DG formation and is required for the maturation of granule cells during DG development30. Next, we assayed the binding of three histone markers on Prox1. The results showed decreases in H3K4me3 and H3K27ac and an increase in the H3K27me3 marker at the promoter region of Prox1(Fig. 6E). We also found BRG1 has binding on the Prox1 promoter (Fig. 6E). Consistent with this, the expression level of Prox1 was downregulated in cKO mice. Therefore, these results indicate Arid1a mediates BAF functions to establish the poised chromatin configuration, which is essentially the proper DG development.
Overexpression of Prox1 rescues the proliferation and differentiation defects of NSPCs in Arid1a cKO hippocampus
To examine a functional relationship between Arid1a and Prox1 in mediating neural development, we then examined the expression of Prox1. Immunostaining results showed that the expression of Prox1 is indeed reduced in the DG of Arid1a cKO mice at prenatal and postnatal stages (Fig. 7A, B). To determine whether Arid1a regulates Prox1 expression directly, we chose 3000 base pairs(bp) promoter according to reference and cloned every 1000bp of the Prox1 promoter. The dual-luciferase reporter assay suggested that ARID1A was bound to Prox1 promoter-1(Supplemental Fig. 6A, B).
Given the substantial decrease of Prox1 in the hippocampal upon the loss of Arid1a, we thus reasoned whether Prox1 gain-of-function could ameliorate the defects of Arid1a cKO hippocampal development. To test this, we first constructed Prox1 overexpression virus and infected cultured Arid1a cKO hippocampal NSPCs and assessed the impact of Prox1 overexpression on the abnormal proliferation and differentiation phenotypes associated with the loss of Arid1a. Western blotting showed increased PROX1 protein levels in the Prox1 overexpression group (Supplemental Fig. 6C). Prox1 overexpression could reverse the reduced NSPCs proliferation and differentiation induced by Arid1a deletion measured by the number of BrdU+GFP+ cells (Fig. 7C, D) and the number of Tuj1+GFP+ cells (Fig. 7E, F). These results suggest that Prox1 is a functional downstream target of Arid1a in modulating NSPCs proliferation and differentiation.