Skip to main content
PLOS Biology logoLink to PLOS Biology
. 2024 Sep 3;22(9):e3002760. doi: 10.1371/journal.pbio.3002760

Phosphorylation of the DNA damage repair factor 53BP1 by ATM kinase controls neurodevelopmental programs in cortical brain organoids

Bitna Lim 1,¤,#, Yurika Matsui 1,#, Seunghyun Jung 1, Mohamed Nadhir Djekidel 2, Wenjie Qi 2, Zuo-Fei Yuan 3, Xusheng Wang 3, Xiaoyang Yang 1, Nina Connolly 1, Abbas Shirinifard Pilehroud 1, Haitao Pan 4, Fang Wang 4, Shondra M Pruett-Miller 5, Kanisha Kavdia 3, Vishwajeeth Pagala 3, Yiping Fan 2, Junmin Peng 1,6, Beisi Xu 2, Jamy C Peng 1,*
Editor: Madeline Lancaster7
PMCID: PMC11398655  PMID: 39226322

Abstract

53BP1 is a well-established DNA damage repair factor that has recently emerged to critically regulate gene expression for tumor suppression and neural development. However, its precise function and regulatory mechanisms remain unclear. Here, we showed that phosphorylation of 53BP1 at serine 25 by ATM is required for neural progenitor cell proliferation and neuronal differentiation in cortical brain organoids. Dynamic phosphorylation of 53BP1-serine 25 controls 53BP1 target genes governing neuronal differentiation and function, cellular response to stress, and apoptosis. Mechanistically, ATM and RNF168 govern 53BP1’s binding to gene loci to directly affect gene regulation, especially at genes for neuronal differentiation and maturation. 53BP1 serine 25 phosphorylation effectively impedes its binding to bivalent or H3K27me3-occupied promoters, especially at genes regulating H3K4 methylation, neuronal functions, and cell proliferation. Beyond 53BP1, ATM-dependent phosphorylation displays wide-ranging effects, regulating factors in neuronal differentiation, cytoskeleton, p53 regulation, as well as key signaling pathways such as ATM, BDNF, and WNT during cortical organoid differentiation. Together, our data suggest that the interplay between 53BP1 and ATM orchestrates essential genetic programs for cell morphogenesis, tissue organization, and developmental pathways crucial for human cortical development.


53BP1 is a DNA damage repair factor that regulates gene expression for tumor suppression and neural development, but its precise regulatory mechanisms are unclear. This study shows that phosphorylation of 53BP1 by ATM kinase is crucial for modulating genetic programs in neural progenitors and cortical organoids.

Introduction

Transcription ensures the proper expression of genetic information for the development and function of the organism, whereas DNA repair maintains the integrity of the genetic code. These 2 processes share cross-functional factors, including CSB, TFII, and XPG, which repair DNA damage caused by torsional stress from transcription-initiating RNA polymerase II [13]. Conversely, some proteins initially believed to function exclusively in DNA repair have been found to regulate gene expression. For example, 53BP1 (p53 binding protein 1) is a key regulator of DNA repair mechanisms, promoting nonhomologous end-joining over homologous recombination [4]. During the DNA damage response, 53BP1 plays a pivotal role in p53-mediated activation of tumor suppressive genetic programs [5]. Recent research has also revealed that 53BP1 collaborates with the chromatin modifier UTX in neural progenitor cells (NPCs), promoting an open chromatin to facilitate the activation of neurogenic or corticogenic programs [6]. Intriguingly, the 53BP1–UTX interaction is observed in humans but not in mice [6]; the mechanism is not well conserved and regulates primate neurodevelopment. These discoveries highlight the importance of 53BP1 in gene regulation for tumor suppression and neural development. However, the precise mechanisms underlying 53BP1’s role in gene regulation and its upstream mechanism are yet to be fully understood.

Studies of 53BP1 have primarily focused on its role in the DNA damage response. To localize to chromatin with double-stranded breaks, 53BP1 uses its BRCT domain to bind to γH2AX, the Tudor domain to bind to H4K20 dimethylation, and its UDR segment to bind to ubiquitinated H2AK15 [710]. Additionally, the phosphorylated SQ/TQ motif of 53BP1 coordinates the docking of RIF1 or SCAI, selectively promoting nonhomologous end-joining or reducing homologous recombination [11,12]. These interactions are likely relevant to the gene regulatory activities of 53BP1. For example, γH2AX recruits 53BP1 and is required for resolving R-loops, DNA demethylation, transcription activation, and transcription elongation [13,14]. These findings suggest that the activities of 53BP1 in DNA damage response are interconnected with its gene regulatory functions.

The studies mentioned above have contributed to a model of 53BP1, where posttranslational modifications of its different residues and domains coordinate various activities. Most prominently, numerous residues of 53BP1 are phosphorylated by ATM (ataxia telangiectasia mutated) kinase [10,1517]. ATM-mediated phosphorylation of 53BP1 or 53BP1-interacting proteins controls protein interactions, cellular localization, and DNA repair mechanisms [11,12,1820]. Despite these discoveries, the impact of phosphorylation on the gene regulatory activity of 53BP1 remains unknown. Here, we report that phosphorylation of 53BP1-serine 25 by ATM is crucial for the proper expression of genetic programs during the growth and development of cortical brain organoids. ATM-dependent phosphorylation controls the chromatin binding of 53BP1 to genomic targets functioning in several key pathways, including neuronal differentiation, cytoskeleton, p53, and ATM, BNDF, and WNT signaling pathways. These results highlight the essential role of 53BP1 phosphorylation in regulating genetic programs for the differentiation of cortical brain organoids.

Results

Phosphorylated 53BP1-S25 increases during differentiation of hESCs into NPCs

Although 53BP1 is required for human embryonic stem cells (hESCs) to differentiate into NPCs [6], its levels did not change during differentiation (Fig 1A). Therefore, its regulation is likely posttranslational during neural differentiation. Human NPCs were analyzed by RNA-seq and immunofluorescence to validate successful NPC generation (S1A-S1E Fig). 53BP1 is targeted by various kinases, including ATM, and we hypothesized that 53BP1 phosphorylation regulates the differentiation of hESCs into NPCs. Intriguingly, we found that the levels of 53BP1 phosphorylated at serine 25 (53BP1-pS25) were markedly increased in NPCs compared to hESCs (Figs 1A and S1F-S1H). The levels of the DNA damage marker γH2AX were similar in NPCs and hESCs (S1I Fig), suggesting that the increase in 53BP1-pS25 levels during NPC differentiation is not due to increased DNA damage.

Fig 1. ATM binds 53BP1, is required for pS25-53BP1, and promotes cortical organoid differentiation.

Fig 1

(A) WB of the nuclear extract of hESCs and hNPCs showed marked increase of 53BP1-pS25 in hNPCs. WB analysis of IgG, (B) 53BP1, and (C) ATM co-immunoprecipitation in the nuclear extract of hESCs. (D) Quantification of the relative ATM protein levels (normalized to β-ACTIN) in 5 replicate WB analyses of hESCs and hNPCs. (E) Schematic diagram of the cortical organoid differentiation. Aggregates were formed in the induction media for 17 days, embedded in Matrigel droplets and cultured in cortical differentiation medium for 16 days, and then cultured in cortical maturation media thereafter. (F) WB analysis of WT and ATM-KO cortical organoids at day 35 of differentiation. Immunofluorescence of (G) PAX6 and CTIP2 and (J) KI67 in cryosections of cortical organoids at day 35 of differentiation. Bar, 100 μm. At day 35 of differentiation, the (H) area and (I) thickness of VZ-like regions were compared between groups. Data points represent single organoids. The mean ± SEM values were compared by one-way ANOVA with Dunnett’s multiple comparisons test to yield **** indicating p < 0.0001. n = 13 organoids/group. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; DMEM, Dulbecco’s Modified Eagle Medium; GMEM, Glasgow Modified Essential Medium; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; IgG, immunoglobulin G; KO, knockout; KSR, Knockout Serum Replacement; VZ, ventricular zone; WB, western blot; WT, wild type; 53BP1, p53 binding protein 1; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

ATM is required for 53BP1-S25 phosphorylation, cell differentiation, and tissue morphogenesis in cortical organoids

The ATM kinase phosphorylates 53BP1-S25 [15], and we thus investigated whether ATM plays a role in neural differentiation. First, we found that ATM co-immunoprecipitated with 53BP1, as did the positive control UTX, but not with the negative control SUZ12 (a core subunit of PRC2, which does not bind these proteins; Fig 1B). Similarly, 53BP1 co-immunoprecipitated with ATM, but not with the negative control SUZ12 (Fig 1C). Like 53BP1-pS25, ATM levels were significantly increased in NPCs compared with hESCs (Fig 1D). ATM up-regulation in NPCs was shown by a previous DNA damage response study [21].

Next, we used the CRISPR-Cas9 system to generate 4 ATM-knockout (KO) hESC lines (Figs 2A, 2B, and S1J). Data from RNA-seq and immunofluorescence showed that ATM-KO did not markedly alter hESC pluripotency (S2C and S2D Fig). All cell lines underwent karyotyping analysis and were characterized as karyotypically normal (S1 Table). ATM-KO lines had minor abnormalities, as expected due to the requirement of ATM for DNA damage repair. To analyze the role of ATM in human cortical development, we used an established protocol to differentiate wild type (WT) and ATM-KO hESCs into cortical organoids (Fig 1E, Methods; [6]). We did not detect 53BP1-pS25 in ATM-KO D35 cortical organoids (Fig 1F) nor NPCs (S2B Fig), consistent with loss of ATM-mediated phosphorylation of 53BP1-S25 during neural differentiation of hESCs. ATM-KO modestly reduced γH2AX levels in NPCs (S2E Fig), suggesting that ATM promotes the phosphorylation of H2AX-S139 in NPCs.

Fig 2. Analysis of neuronal differentiation and cell organization in cortical organoids.

Fig 2

Quantification of PAX6/CTIP2 ratios in (A) D28 and (B) D37 cortical organoids. (C) Quantification of NEUN/DAPI in D37 cortical organoids. (D) In D37 cortical organoids, 6 organoids were surveyed to count ZO-1-positive apical surfaces and proportions of PAX6-positive NPCs that are organized around the apical surfaces. (E) Proportions of PH3-positive cells that are adjacent to ZO-1-positive apical surfaces (“rings”). Data from 53BP1-S25A and S25D were included for comparison. **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; ns, not significant by Welch’s t test in (A-C) and two-way ANOVA test in (D). From GSEA, functional terms that are highly enriched in (F) up-regulated and (G) down-regulated genes in ATM-KO vs. WT NPCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type; 53BP1, p53 binding protein 1.

By day 35 (D35) of differentiation, WT cortical organoids expressed the forebrain NPC marker PAX6 in ventricular zone–like regions that were radially organized (Fig 1G). In contrast, ATM-KO D35 cortical organoids displayed disorganized and smaller ventricular zone–like regions (Fig 1G-1I). We quantitatively compared NPC proliferation, neuronal differentiation, cell death, and cell organization in ATM-KO versus WT cortical organoids. Examination of endogenous DNA damage, by γH2AX immunofluorescence, did not reveal marked difference (S3A Fig), confirming our western blot (WB) results in S1I Fig. Although quantification of cell death marker cleaved-caspase 3 by FACS revealed a modest increase of cell death in D21 ATM-KO cortical organoids, FACS and immunofluorescence quantification showed that D28 and D35 ATM-KO and WT cortical organoids are similar in cell death frequencies (S3B-S3E Fig). Cell proliferation frequencies did not significantly differ between D28 and D35 ATM-KO and WT cortical organoids (S4 Fig). We next quantified immature neuronal marker NEUN and PAX6/CTIP2 ratios. Despite lower levels of immature neuronal differentiation, ATM-KO exhibited higher neuronal maturation (Figs 2A-2C and S5A). These data suggest that ATM-KO fastens the phase of immature neuronal differentiation, leading to enhanced neuronal maturation. Finally, we quantified ZO-1-positive ventricular surfaces and the organization of PH3-positive and PAX6-positive cells around ventricular surfaces. The ATM-KO ventricular surfaces were similar to WT at D28 (S5B-S5D Fig), but the number was much reduced by D37 (Fig 2D). NPC organization around the ventricular surfaces were similarly organized in D37 (Figs 2D and S5E); however, fewer ATM-KO proliferative cells were adjacent to ventricular surfaces (Fig 2E). These data suggest that ATM-KO enhances neuronal maturation and cellular disorganization in developing cortical organoids. By D55, ATM-KO cortical organoids had similar size distribution as the control (S5F and S5G Fig). Thus, ATM controls neuronal differentiation and cellular organization to form ventricular zone–like regions in cortical organoids.

ATM safeguards transcriptional and translational programs in differentiating cortical organoids

To investigate the molecular basis of the cellular defects we observed in ATM-KO, we performed RNA-seq to compare ATM-KO to WT NPCs and D35 cortical organoids derived from WT and ATM-KO hESCs. The expression of forebrain markers was similar between WT and ATM-KO cortical organoids (and low expression of midbrain and hindbrain markers; S2 Table), suggesting that the ATM-KO cortical organoids specified to the forebrain lineage. A false discovery rate (FDR) <0.05 was used to identify differentially expressed genes. Gene set enrichment analysis (GSEA) showed that up-regulated genes in ATM-KO NPCs were enriched in forebrain development, axis specification, and metabolic pathways (Figs 2F and S6A), whereas down-regulated genes were enriched in neuronal differentiation, epithelial mesenchymal transition, and tube morphogenesis (Fig 2G). Comparison of transcriptomes profiles of D35 cortical organoids from 8 ATM-KO versus 6 WT datasets yielded similar GSEA terms (Fig 3A and 3B). These data suggest that ATM regulates genetic programs related to forebrain development, metabolism, and neuronal differentiation in NPCs and cortical organoids. Dysregulated genetic programs likely contributed to enhanced neuronal maturation and cellular disorganization in ATM-KO cortical organoids.

Fig 3. Transcriptomic and proteomic profiles of WT versus ATM-KO cortical organoids.

Fig 3

From GSEA, functional terms that are highly enriched in (A) up-regulated and (B) down-regulated genes in ATM-KO D35 cortical organoids. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. (C) Schematic diagram outlining TMT LC-MS/MS profiling of total proteomics and phosphoproteomics of D35 WT and ATM-KO cortical organoids. TMT signals from total proteomics were used to normalize those of phosphopeptides. (D) Using FC>1.5 and FDR<0.05, 198 phosphoproteins were found to be lower in 2 ATM-KO versus WT. (E) Normalized levels of phosphoproteins that have ATM-dependent phosphorylation in D35 cortical organoids. 53BP1 and EIF4EBP1 were known substrates of ATM. The error bars depict the mean and standard error of the mean values, which were calculated based on the normalized levels of each phosphopeptide in the protein. (F) Enrichment of proteins with ATM-dependent phosphorylation in specific functional categories. (G) Heatmap showing altered activities of kinases between D35 ATM-KO2 and WT cortical organoids. Relative changes in kinase activity are shown as row Z‐scores. Kinase activity was inferred by IKAP [25] based on normalized substrate phosphorylation levels from phosphor-proteome. The normalization was performed by dividing phosphor-peptide abundance of each protein by corresponding protein abundance [57]. Circos plots showing kinases with inferred (H) higher and (I) lower activities in D35 ATM-KO versus WT cortical organoids and their corresponding enriched pathways. Underlying numerical values for figures are found in S1_Data.xlsx. ATM, ataxia telangiectasia mutated; FC, fold-change; FDR, false discovery rate; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; TMT LC-MS/MS, liquid chromatography-tandem mass spectrometry; WT, wild type; 53BP1, p53 binding protein 1.

As ATM kinase is crucial for many cell and developmental processes, we aimed to analyze its effect on the proteome and phosphoproteome of differentiating cortical organoids. First, we used multiplexed tandem mass tag-based quantification and 2D liquid chromatography-tandem mass spectrometry (TMT LC-MS/MS) to profile the proteome of WT and ATM-KO D35 cortical organoids (S3B Fig, Methods). We quantified 10,895 proteins between 4 WT, 4 ATM-KO2, 3 ATM-KO3, and 3 ATM-KO14 D35 cortical organoid samples by using the criteria of fold change >1.5 and FDR <0.05 (Fig 3C). Consistency between replicate datasets is supported by principal component analysis (S3B Fig). GSEA showed that compared to WT, up-regulated proteins in ATM-KO were enriched in terms related to neurotransmission, neuron spine, dendrite, synapse, and axon (S3C Fig), whereas down-regulated proteins were enriched in BMP/TGFβ and WNT signaling, epithelial morphogenesis, and stem cell differentiation (S3D Fig). These data suggest that ATM controls posttranscriptional and translational gene regulation to suppress neuronal function and promote stem cell differentiation, epithelial morphogenesis, and TGFβ and WNT signaling pathways in D35 cortical organoids.

We have observed distinct patterns in the transcriptomics and proteomics data in ATM-KO versus WT cortical organoids. Interestingly, while transcriptomic programs related to neuronal differentiation were down-regulated (Fig 3B), proteomic programs related to neuronal function were up-regulated (S3C Fig) in ATM-KO versus WT cortical organoids. These differential patterns in transcriptomics and proteomics are likely a consequence of the regulatory role of ATM in multiple cellular processes. It is possible that the higher protein expression related to neuronal functions in ATM-KO lead to down-regulation of transcriptional expression of neuronal differentiation programs. This would suggest that the dysregulated transcriptomic and proteomic programs in ATM-KO cortical organoids are interconnected and result from the complex interplay of ATM’s regulation of various cellular pathways. These findings shed light on the intricate role of ATM in coordinating gene expression and protein levels, influencing neuronal differentiation and function in cortical organoids.

ATM-dependent phosphorylation controls signaling pathways for neurogenesis, stem cell differentiation, and morphogenesis in cortical organoids

To investigate how ATM exerts its modulatory control during cortical organoid formation, we performed phosphoproteomics analysis of WT and ATM-KO cortical organoids. Using TMT LC-MS/MS, we quantified 22,646 phosphopeptides and normalized their abundance based on the protein abundance measured in the total proteomics analysis. A comparison between WT and ATM-KO lines revealed that 198 proteins had consistently lower levels of phosphorylation in at least 2 of the 3 ATM-KO lines (log2(fold change >1.5) and FDR <0.05; Fig 3D and S3 Table). Among these proteins, 53BP1 and EIF4EBP1 were known substrates of ATM (Fig 3E) [15,16,22,23], validating the approach to identify putative ATM substrates in cortical organoids. However, it is essential to note that this approach does not distinguish between direct and indirect effects, and, therefore, some of the identified proteins could be phosphorylated by protein kinases that require ATM for their activity. Notably, many ATM-dependent phosphorylated proteins were found to be key neurodevelopmental regulators (Fig 3E) and enriched in functions related to neurodevelopment, neurogenesis, cell morphogenesis, and cytoskeleton (Fig 3F). These findings suggest that ATM plays a critical role in regulating the phosphorylation of proteins involved in essential processes for neurodevelopment and neuronal function in cortical organoids.

We further explored the effects of ATM by identifying protein kinases that had ATM-dependent phosphorylation. We used the IKAP machine learning algorithm [24] to analyze substrates (inferred from literature curation) and deduce the activities of those kinases. For example, in ATM-KO compared to WT, we found reduced phosphorylation of proteins related to MAPK9 activities, such as DCX, MAPT, and NFATC4 (S7A Fig and S4 Table) [24]. On the other hand, we found higher phosphorylation of proteins related to CDK5 activities, including ADD2, ADD3, DCX, DNM1L, DPYSL3, MAPT, and SRC (S7B Fig and S4 Table) [24]. In ATM-KO, we inferred lower activities in MAPK9, CDK2, CHEK1, ATR, CSNK1A1, MTOR, CAMK2A, and PRKACA (Figs 3G and S7C), with enriched functions in ATM signaling, BNDF signaling, and axon guidance (Figs 3G, 3H, and S7C). On the other hand, we inferred higher activities in GSK3B, MAPK3, PAK1, CSNK2A1, CDK5, CDK1, and PRKDC (Figs 3G, 3I, and S7C), with enriched function in ATM signaling, WNT signaling, G2/M checkpoint, and p53 regulation in ATM-KO (Fig 3H and 3I). ATM KO leads to both lower and higher activities of kinases in ATM signaling. Additionally, some of the altered kinase activities could be secondary to ATM-KO, as CHEK1, ATR, and PRKDC were known substrates of ATM [23,25]. Overall, these data suggest that the activities of kinases related to ATM signaling, BNDF signaling, WNT signaling, G2/M checkpoint, and p53 regulation became dysregulated in ATM-KO D35 cortical organoids.

We thus conclude that ATM plays a crucial role in controlling key neurodevelopmental regulators. The dysregulated phosphorylation and activities of these regulators disrupt the normal transcriptomic program responsible for neuronal differentiation, leading to higher proteomic programs associated with neuronal function. As a consequence, the dysregulated programs in ATM-KO cortical organoids are likely responsible for the observed defects in neurogenesis and morphogenesis (formation of ventricular zone–like regions). These findings provide valuable insights into the role of ATM in neurodevelopment and shed light on potential molecular mechanisms underlying neurological disorders associated with ATM dysfunction.

Phosphorylation of 53BP1-S25 coordinates NPC proliferation and neuronal differentiation

We next examined ATM-dependent phosphorylation of 53BP1-S25. To specifically investigate the functional significance of 53BP1-pS25, we used the CRISPR-Cas9 system to mutate the endogenous 53BP1 serine 25 to alanine (S25A) or aspartic acid (S25D) (Figs 4A and S7D, Methods). The alanine substitution precludes phosphorylation, whereas aspartic acid is chemically similar to phosphoserine [26]. We generated 4 53BP1-S25A hESC lines (34–3, 34–4, 79–1, and 79–3) and 4 53BP1-S25D hESC lines (14–3, 14–15, 14–19, and 17). The total levels of 53BP1 were similar in WT, 53BP1-S25A, and 53BP1-S25D NPCs, and we did not detect pS25 in 53BP1-S25A NPCs, as expected (S1D and S7E Figs). Control, 53BP1-S25A, and 53BP1-S25D hESC lines displayed similar transcriptomic profiles and pluripotency marker expression (S2C, S7F, and S8A Figs), suggesting that 53BP1-S25A and 53BP1-S25D do not affect hESC self-renewal.

Fig 4. 53BP1-pS25 is required for the differentiation of cortical organoids.

Fig 4

(A) In the endogenous 53BP1 locus, the codon TCT encoding serine-25 in was mutated to GCT and GAT encoding alanine and glutamate, respectively. (B) Bright-field images of cortical organoids formed by 4 53BP1-S25A lines, 4 53BP1-S25D lines, and 2 WT control at day 35 of differentiation. Bar, 1.5 mm. At day 35 of differentiation, the (C) organoid size and (F) area of ventricular zone–like region were compared between groups. Data points represent single organoids. The mean ± SEM values were compared by one-way ANOVA with Dunnett’s multiple comparisons test to yield ****, ***, **, *, and ns indicating p < 0.0001, 0.001, 0.01, 0.05, and not significant, respectively. n = 39–47 organoids/group for (C) and 15–33 organoids/group for (F). (D) Immunofluorescence of PAX6 and CTIP2 in cryosections of cortical organoids at day 35 of differentiation. Bar, 100 μm. (E) Illustration of ventricular zone–like areas in cortical organoids. Underlying numerical values for figures are found in S1 Data. WT, wild type; 53BP1, p53 binding protein 1; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

ATM is required for the phosphorylation of many neurodevelopmental regulators (Fig 3F). As the role of 53BP1-S25 beyond DNA damage repair is not known, we seek to analyze its role in human cortical development. We differentiated control WT, 53BP1-S25A, and 53BP1-S25D hESCs into cortical organoids (Fig 1E, Methods). The 53BP1-S25A and 53BP1-S25D D35 cortical organoids displayed smaller sizes compared to WT controls (Fig 4B and 4C), suggesting that phosphorylation at S25 is essential for cortical organoid growth and development. Analysis of the ventricular zone–like regions showed 53BP1-S25A and 53BP1-S25D are significantly smaller than those in WT (Fig 4D-4F). Fewer cells were positive for KI67 (proliferation marker) or phosphorylated-serine 10 histone H3 (mitotic chromatin marker) (S8B–S8D Fig). Examination of endogenous DNA damage and cell death, assessed by γH2AX and cleaved-caspase 3, respectively, did not reveal significant differences between 53BP1-S25A, S25D, and WT (S9 Fig). To explore the developmental timing of the cellular phenotypes, we quantified KI67, NPC marker PAX6, and neuronal marker CTIP2 in D14, D21, D28, and D35 cortical organoids. At D28, 53BP1-S25A and S25D cortical organoids had significantly lower cell proliferation and higher neuronal differentiation (Fig 5). Quantification of the tight junction protein ZO-1 showed significantly fewer ZO-1-positive ventricular surfaces in D28 53BP1-S25A and S25D cortical organoids compared to WT (S10 Fig). For the ventricles that did form in D28 53BP1-S25A and S25D, their surface areas did not significantly differ from those of WT (S10B Fig). These data suggest that lower ventricle formation, lower cell proliferation, and higher neuronal differentiation contributed to the depletion of progenitor pools and smaller cortical organoids in 53BP1-S25A and S25D.

Fig 5. 53BP1-S25A and S25D lower cell proliferation in cortical organoids.

Fig 5

FACS quantified ratios of (A) KI67, (B) PAX6, and (C) CTIP2 to total cells in D28 cortical organoids. (D) Immunofluorescence of PAX6 and CTIP2 in D28 cortical organoids. Bar, 100 μm. Quantification of immunofluorescence signals of (E) PAX6/DAPI, (F) CTIP2/DAPI, and (G) PAX6/CTIP2 in D28 cortical organoids. Each data point represents quantification of cells in 1 cortical organoid. Quantification of (H) KI67/PAX6 and (I) PAX6/CTIP2 ratios in immunofluorescence of D35 cortical organoids. Each data point represents quantification of cells in 1 cortical organoid. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data.

At D55, the 53BP1-S25A and S25D cortical organoids remained significantly smaller than WT (S12 Fig and S5 and S6 Tables). These data suggest that the cell biological effects of the S25A and S25D mutations were similar, despite the aspartic acid mutation (S25D) being chemically similar to phosphoserine, which is the phosphorylated form of S25. The S25D mutation may act as an inhibitory mimic of phosphorylation, akin to the S25A mutation. Consequently, the absence of S25 phosphorylation impacts NPC proliferation and overall cortical organoid growth.

Phosphorylation of 53BP1-S25 modulates the expression of genetic programs for neuronal differentiation and function

Using RNA-seq, we examined the transcriptomes of WT (6 samples), 53BP1-S25A (8 samples), and 53BP1-S25D (8 samples) D35 cortical organoids. We analyzed expressed genes with counts per million values >1 and observed few differences in gene expression between 53BP1-S25A and 53BP1-S25D D35 cortical organoids, using FDR <0.05 (Fig 6A). When comparing the transcriptomes of 53BP1-S25A and 53BP1-S25D organoids to WT, there were high concordant changes in gene expression, with over 87% of differentially expressed genes in 53BP1-S25A also being altered in 53BP1-S25D (Figs 6B and S12D). However, 53BP1-S25D disrupted the expression of 2- to 3-fold more genes than 53BP1-S25A, suggesting a gain-of-function effect for the 53BP1-S25D mutation. To explore this further, we performed GSEA and found that the top terms enriched in the up-regulated genes of 53BP1-S25A and S25D organoids were highly overlapping (Fig 6C). In contrast, there was low overlap of the top terms in the down-regulated genes in 53BP1-S25A versus WT and 53BP1-S25D versus WT (S12E Fig). Both mutations led to the up-regulation of genes related to synapse, axon, and neurotransmitter functions, suggesting a shared effect on enhancing neuronal function (Fig 6C). The S25D mutation specifically up-regulated more genes involved in neuronal function compared to S25A, indicating a stronger impact on this aspect of gene regulation (Fig 6C and 6D). These findings highlight the significance of the S25 phosphorylation site in 53BP1 for the regulation of genes involved in neuronal function and support that the S25D mutation results in a gain-of-function effect, leading to more pronounced changes in gene expression related to neuronal processes.

Fig 6. 53BP1-S25 phosphorylation enforce the appropriate expression of genetic programs for cortical organoid differentiation.

Fig 6

(A) Number of differentially expressed genes identified by pairwise comparisons at FDR <0.05. At day 35 of differentiation, 53BP1-S25A and S25D cortical organoids are molecularly similar. (B) Differentially expressed genes in 53BP1-S25D versus WT overlap 87% (764/875) and 91% (361/396) of those in 53BP1-S25A versus WT. (C) Extensive overlap of up-regulated GSEA terms between 53BP1-S25A versus WT and 53BP1-S25D versus WT. Most terms relate to axon, synapse, and neurotransmitter. (D) Of 53BP1 target genes up-regulated by S25A and S25D, 212 genes require WT 53BP1 for expression in cortical organoids. (E) The 212 genes are enriched in functions related to transcriptional regulation, neuron projection, axonogenesis, synapse, neurotransmitter synthesis and transport, and membrane depolarization. (F) Venn diagrams depict high overlaps between down-regulated genes in all 3 groups of mutant versus WT pairwise comparisons. (G) GSEA graphs showed that down-regulated genes in 53BP1-S25A or S25D vs. WT had significant enrichment in down-regulated genes of ATM-KO vs. WT cortical organoids. P values were calculated by the hypergeometric test, assuming normal data distribution. (H) GSEA terms of the 115 genes that were down-regulated in all 3 groups (versus WT) revealed the genetic programs copromoted by ATM and 53BP1-pS25. ATM, ataxia telangiectasia mutated; FDR, false discovery rate; GSEA, gene set enrichment analysis; KO, knockout; WT, wild type; 53BP1, p53 binding protein 1.

It remained unclear whether the higher expression of neuronal genetic programs in the 53BP1 mutants occurred in NPCs or neurons. Therefore, we compared the transcriptomes of 53BP1-S25A and S25D to WT NPCs, which had similar expression of NPC markers PAX6 and NES (S12A Fig). Up-regulated genetic programs in 53BP1-S25A and S25D NPCs shared categories such as translation control and ribosome (S12B and S12C Fig), whereas 53BP1-S25A NPCs also up-regulated cell cycle control and chromosome segregation (S12C Fig). Surprisingly, down-regulated genetic programs in 53BP1-S25A and S25D NPCs were highly enriched in neuronal differentiation (S12D and S12E Fig). The down-regulated genetic programs in NPCs are similar to neuronal programs that became up-regulated in 53BP1-S25A and S25D versus WT cortical organoids. These data suggest that 53BP1-S25 phosphorylation promotes the appropriate expression of neurogenic programs in NPCs and modulates the expression of the same programs in differentiating neurons in cortical organoids.

To dig deeper into analyses, we compared our data with previously published transcriptomic data that compared 53BP1-KO and WT cortical organoids, which support a requirement of 53BP1 for activating neurogenic genes [6]. We observed that gene categories up-regulated by 53BP1-S25A and S25D were similar to those down-regulated in 53BP1-KO cortical organoids. This was a significant overlap of 212 genes up-regulated by 53BP1-S25A and 53BP1-S25D with 53BP1-bound target genes that were down-regulated in 53BP1-KO versus WT (p = 0 by empirical estimation; Fig 6D). The 212 genes were enriched in functions related to regulation of transcription, neurogenesis, neuronal projection, axonogenesis, synapse organization, and membrane depolarization (Fig 6E). This suggests that the expression of these genes is dependent on and up-regulated by 53BP1 phosphorylated at S25 in cortical organoids.

We next examined how transcriptomic changes in 53BP1-S25A and S25D compared to those in ATM-KO cortical organoids. We observed little overlap between the down-regulated genes in ATM-KO and the up-regulated genes in 53BP1-S25A and 53BP1-S25D. In contrast, we observed a greater overlap in concordant gene expression changes in ATM-KO, 53BP1-S25A, and 53BP1-S25D versus WT (Figs 6F, S12F, and S12G). GSEA showed a significant enrichment of concordantly differentially expressed genes among ATM-KO, 53BP1-S25A, and 53BP1-S25D versus WT (Figs 6G and S13A-S13C). Notably, all 3 mutant types shared down-regulated genes that were enriched functions related to TNFα signaling via NFκB, p53 pathway, IRE1-mediated unfolded protein response, FGFR signaling, TGFβ signaling, apoptosis, regulation of cell proliferation, and epithelial mesenchymal transition (Fig 6G). These data suggest that both ATM and 53BP1-pS25 promote the expression of these genes. From these findings, we can infer that ATM likely promotes the expression of these genes via phosphorylating 53BP1 at S25 in D35 cortical organoids. This suggests that ATM and 53BP1 may function together in a coordinated manner to regulate the expression of genes involved in critical signaling pathways and cellular processes during cortical development.

53BP1-S25A and S25D predominantly alter the expression of 53BP1 target genes

To obtain further mechanistic insights into the role of 53BP1 in controlling gene expression, we reanalyzed 53BP1 ChIP-seq data (using 2 separate anti-53BP1 antibodies) in WT NPCs [6]. Using SICER [27] and MACS2 [28] with a criterion of FDR <0.05, we identified 37,519 targets bound by 53BP1. About 41% of these 53BP1 targets localize to promoter regions, suggesting a transcriptional regulatory role of 53BP1 (S14D Fig). Remarkably, more than 82% of the differentially expressed genes in 53BP1-S25A and 53BP1-S25D D35 cortical organoids were found to be targets bound by 53BP1 (Figs 7A, S13E, and S13F). 53BP1 target genes with increased transcript levels in the mutant organoids were highly enriched in neuronal development, axonogenesis, neuron projection, synapse organization, and neurotransmitter transport, transmission, and signaling (Fig 7B). On the other hand, 53BP1 targets with reduced transcript levels in the mutant organoids were enriched in IRE1-mediated unfolded protein response, cellular response to stress, iron import, and apoptosis regulation (S13G Fig). Of note, genes involved in IRE1-mediated unfolded protein response and apoptosis regulation showed reduced expression upon loss of ATM or mutation of 53BP1-S25 and were identified as direct targets of 53BP1 in NPCs. This suggests that ATM-mediated phosphorylation of 53BP1-S25 directly promotes the expression of these genes to maintain NPCs during formation of cortical organoids.

Fig 7. 53BP1-pS25 positively and negatively regulate 53BP1 target genes.

Fig 7

(A) More than 82% of differentially expressed genes in 53BP1-S25A or S25D versus WT are chromatin targets bound by 53BP1 in WT NPCs. (B) 53BP1-S25A and S25D up-regulate 53BP1 targets that are involved in neuron development and projection, axonogenesis, synapse, and neurotransmitter synthesis and transport. (C) Heatmaps aligning peaks with 53BP1-pS25 CUT&RUN and 53BP1 ChIP-seq signals in WT NPCs. Input track was included as a negative control. n = numbers of peaks with differential and overlapped bindings. Criteria of FC>2 and p < 0.05 were used for comparison. (D) GSEA graph of 53BP1-pS25 CUT&RUN signals in genes that were lower in ESCs vs. NPCs, which were up-regulated in NPCs. P values were calculated by the hypergeometric test, assuming normal data distribution. Heatmaps aligning peaks with significantly different 53BP1 ChIP-seq signals in (E) 53BP1-S25A vs. WT and (F) 53BP1-S25D vs. WT, using the criterion of FC>2 and p < 0.05. Control peaks are those, after voom normalization, showed the least changes and served as semi-independent validation of differential ChIP-seq analysis. Bubble graphs present top enriched categories of genes that had significantly higher 53BP1 ChIP-seq in (G) 53BP1-S25A vs. WT and (H) 53BP1-S25D vs. WT. ESC, embryonic stem cell; FC, fold-change; GSEA, gene set enrichment analysis; NPC, neural progenitor cell; WT, wild type; 53BP1, p53 binding protein 1; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

We wanted to test whether ATM alters 53BP1 binding, considering ATM is required for 53BP1-pS25 in D35 cortical organoids (Fig 1F) and NPCs (S2B Fig). A comparison of 53BP1 ChIP-seq in WT and ATM-KO NPCs showed that ATM-KO altered 53BP1 binding to chromatin (S14A-S14C Fig). ATM-KO reduced 53BP1 binding at specific sites, with 96.3% of these sites being promoters (S14C Fig). To explore the impact of 53BP1-pS25, we performed CUT&RUN in 2 separate WT NPC lines. Our analysis revealed that 67.1% of 53BP1-pS25 targets localize to promoter regions, suggesting a transcriptional regulatory role (S14D Fig). Under the criteria of fold-change >2 and p < 0.05, 58.6% (3,390/5,789) of 53BP1-pS25 targets overlapped with 53BP1 targets (Fig 7C); the nonoverlapped sites may be attributed to differences in ChIP versus CUT&RUN procedures and the accessibility of 53BP1 versus 53BP1-pS25 antibodies. 53BP1-pS25 targets were significantly enriched in 414 up-regulated genes in NPCs versus ESCs (Fig 7D), suggesting a role of 53BP1-pS25 in promoting their expression in NPCs. Genes having overlapped 53BP1 ChIP-seq and 53BP1-pS25 CUT&RUN signals were enriched in chromatin remodeling, DNA metabolism, RNA splicing, translation, transcription, cell cycle, and neuron development (S14E Fig). These data suggest that ATM can alter 53BP1 binding and that 53BP1-pS25 is enriched in the promoters of genes regulating cellular processes and neurodevelopment.

Phosphorylation of 53BP1-S25 controls the localization of 53BP1 to chromatin for gene regulation

To investigate the impact of 53BP1-S25 on the genomic distribution of 53BP1, we performed ChIP-seq in 53BP1-WT, S25A, and S25D NPCs. Two independent NPC lines were used for each group, and the ChIP-seq data were subjected to principal component analysis, which showed high consistency between the replicate dataset (S8A Fig). We used SICER [27] and MACS2 [28] with the criteria of fold-change >2 and p < 0.05 to perform pairwise comparisons of the merged datasets from 53BP1-WT, S25A, and S25D ChIP-seq experiments. The pairwise comparisons identified thousands of 53BP1-bound regions that were significantly different between 53BP1-WT, S25A, and S25D. Notably, the regions that significantly gained binding in 53BP1-S25A or S25D versus WT were highly enriched at promoters (within 2 kb of transcription start sites), constituting 82% and 71.1%, respectively (S8B and S8C Fig). In contrast, the regions that significantly lost binding in 53BP1-S25A or S25D versus WT were not as enriched at promoters, constituting 32.6% and 33%, respectively (S8B and S8C Fig). We generated heatmaps to visualize the genomic regions with significantly different 53BP1 binding intensity (compared against control regions). The heatmaps confirmed consistent changes in 53BP1 binding patterns between 53BP1-S25A and S25D versus WT, and between 53BP1-S25A versus S25D (Figs 5C, 5D, and S8D). These data support that 53BP1-S25 and its phosphorylation control the genomic distribution of 53BP1 on chromatin.

We next set out to examine the correlation between changes in 53BP1 distribution on chromatin and changes in gene expression in 53BP1-WT, S25A, and S25D cortical organoids. We performed GSEA and made some notable observations. Firstly, regions that gained 53BP1 binding in 53BP1-S25A or S25D cortical organoids, as compared to WT, were enriched with up-regulated genes (Fig 5E and 5F). Similarly, regions that had lower 53BP1 binding were enriched with down-regulated genes in 53BP1-S25A or S25D versus WT (S8E and S8F Fig). These results suggest that the 53BP1-S25A or S25D mutation directly influences 53BP1 binding and gene expression and subsequently regulates gene expression, particularly at promoters where higher 53BP1 binding leads to higher gene expression.

Interestingly, genes that lost 53BP1-S25A or S25D protein binding had minimal overlap in GSEA terms, except promoters occupied with H3K4me3 and regulation of epithelial-mesenchymal transition (S8E and S8F Fig). In contrast, the genes that gained 53BP1-S25A or S25D protein binding were enriched with promoters marked by bivalent histone marks (H3K4me3 and H3K27me3) or occupied by H3K27me3 alone [29] (Fig 5E and 5F), suggesting that 53BP1-S25D or S25A proteins preferentially bind to these promoters and subsequently up-regulate gene expression. Moreover, genes that gained 53BP1-S25A or S25D binding shared common functions related to sodium ion transmembrane transporter, DNA replication, positive regulation of cell division, and regulation of histone H3K4 methylation (Fig 5E and 5F). This suggests that despite the 1,187 regions showing different 53BP1 bindings between 53BP1-S25A and S25D (S8D Fig), both mutations impact genes involved in neuronal functions and cell proliferation. Altogether, these findings show that 53BP1-S25A and S25D mutations have a direct impact on 53BP1 binding to chromatin and subsequently affecting gene regulation. We propose that 53BP1-pS25 likely inhibits 53BP1 binding to promoters associated with bivalent and H3K27me3-occupied promoters. This inhibition may lead to the reduced expression of genes involved in the regulation of H3K4me3, neuronal functions, and cell proliferation.

Molecular regulation of ATM and 53BP1-pS25 during neural differentiation

We next tried to identify a regulation of ATM, whose protein levels increased in NPCs (Fig 1D). This led us to test whether and how inhibitors of TGFb, WNT, and HH signaling control protein levels of ATM, 53BP1, and pS25-53BP1 by removing one inhibitor at a time from the cortical organoid differentiation media (S16A Fig). As we could not successfully identify physical presence of ATM at promoters, WB analysis is most apt to study ATM level and activity. By day 4 of neural differentiation, although 53BP1 protein levels were reduced by the withdrawal of SB431542 (TGFβ inhibitor) or IWR1-endo (WNT inhibitor), pS25-53BP1 was not altered (S16B and S16C Fig). By day 10 of neural differentiation, the withdrawal of cyclopamine (HH inhibitor) reduced pS25-53BP1 level (but not ATM or 53BP1 proteins; S16D and S16E Fig). These signaling pathways may affect pS25-53BP1 or ATM activities during neural differentiation.

Next, we tested whether another DNA damage response factor, apart from ATM, influences 53BP1-pS25. RNF168 plays a central role in the γH2AX-MDC1-RNF8-RNF168-H2AK15ub axis, which governs the binding of 53BP1 to chromatin with DNA damage [30]. We generated RNF168-KO hESC clone 44, which maintained pluripotency and genome integrity (S17A-S17D Fig and S1 Table). RNF168-KO hESCs were differentiated to NPCs, which expressed NPC markers similar to WT NPCs (S17E Fig). RNA-seq analysis comparing 2 datasets each from RNF168-KO44 and WT NPCs revealed that up-regulated genes were enriched in neuronal differentiation, translation and ribosome, and cell cycle transition (S17F Fig), while down-regulated genes were enriched in cilium movement, H3K27me3 targets, H3K4me3 targets, astrocyte markers, signaling pathways, and positive regulation of NPC proliferation (S17G Fig). We performed 53BP1-pS25 CUT&RUN and showed that RNF168-KO disrupted 53BP1-pS25 localization on chromatin (S17H Fig). RNF168-KO increased 53BP1-pS25 levels at genes enriched in neuronal differentiation, cell morphogenesis, and stem cell maintenance, whereas RNF168-KO decreased 53BP1-pS25 levels at genes enriched in cell cycle transition, signaling receptor regulation, anterior-posterior patterning, and transcription activator (S17I Fig). The altered 53BP1-pS25 localization correlated with differential gene expression in RNF168-KO versus WT NPCs (S17J Fig). Altogether, these data suggest that DNA damage signaling regulates 53BP1 binding to chromatin, affecting genetic programs related to signaling pathways, protein translation, and NPC proliferation and differentiation.”

Discussion

In our study, we made significant discoveries regarding the role of ATM and 53BP1-pS25 in controlling gene expression during the differentiation of hESCs into cortical organoids. We revealed that ATM exerts a strong influence over various aspects of gene regulation, including transcriptional, posttranscriptional, and translational control. While our in vitro model may not fully recapitulate neurodevelopment in vivo, it provides valuable insights into corticogenesis. We have shown that neural differentiation promotes ATM protein levels, and ATM-dependent phosphorylation predominantly impacts factors involved in neurogenesis, neuronal differentiation, cell morphogenesis, and microtubule cytoskeleton. Dysregulation of these processes led to the cellular defects in ATM-KO cortical organoids. We showed that key signaling pathways may affect ATM during neural induction. The activity of ATM can be regulated by DNA damage response, reactive oxygen species, hypothxia, hypothermia, and phosphatase WIP1 [3133]. The exact clarification of mechanisms promoting ATM activities, especially in directing its kinase activity at specific promoters, is beyond the scope of this study. Additionally, we have identified kinases involved in ATM, BDNF, and WNT signaling, G2/M checkpoint, and p53 regulation as being influenced by ATM-dependent phosphorylation during cortical organoid differentiation. These molecular pathways may function in diseases associated with ATM, including ataxia telangiectasia [3436].

We recognized the diverse effects of ATM and decided to focus our studies on 53BP1-pS25, a phosphorylation event dependent on ATM. We found that 53BP1-pS25 regulates genetic programs including signaling pathways, p53 regulation, apoptosis, and cell proliferation. To understand the mechanisms underlying 53BP1’s involvement in gene regulation, we built a model that incorporates current knowledge about 53BP1 functions in the DNA damage response. We propose that ATM phosphorylates H2AX at transcription start sites [13,14], facilitating the recruitment of 53BP1 and subsequent phosphorylation of 53BP1-S25. RNF168, key to DNA damage response signaling [30], also regulates 53BP1-pS25 on chromatin and genetic programs crucial to neural differentiation. Phosphorylation of 53BP1-S25 inhibits the recruitment of 53BP1 to bivalent or H3K27me3-occupied promoters for suppressing the expression of genes involved in the regulation of H3K4me3, neuronal functions, and cell proliferation. The fidelity of gene expression in cortical brain organoids requires dynamic changes in the phosphorylation of 53BP1-S25. This process is likely to involve the interactions of 53BP1 with other proteins, including RIF1, SCAI, and UTX [6,11,12]. These interactors have known roles in chromatin alterations and gene regulation. Notably, UTX is an H3K27me3 demethylase that can modify bivalent or H3K27me3-occupied promoters and has been shown to partner with 53BP1 to promote neurogenesis in humans but not in mice [6]. Given our findings, we propose that 53BP1-pS25 may influence the activities of 53BP1–UTX at bivalent or H3K27me3-occupied promoters, thus modulating gene expression and contributing to the timing of neuronal differentiation.

Our studies have uncovered the remarkable role of ATM–53BP1 in regulating neurodevelopmental programs. Its impact is multifaceted. Firstly, ATM–53BP1 plays a crucial role in maintaining NPCs and controlling the size of cortical organoids. Secondly, ATM–53BP1 is involved in driving and modulating programs related to synapse formation, axon development, and neurotransmitter regulation, processes fundamental for establishing neuronal networks and communication within the brain. Thirdly, our findings reveal a temporal component in the regulation of neurodevelopmental programs by ATM–53BP1. As cortical organoids progress in differentiation, there is a temporal regulation of neuronal differentiation and function. This switch involves ATM and the 53BP1-pS25 dynamics to specifically control genes associated with synapse, axon, and neurotransmitter, which are crucial to cognition. In the future, elucidation of this mechanism will provide valuable insights into the molecular control of corticogenesis. Beyond 53BP1, ATM-dependent phosphorylation likely controls many other key neurodevelopmental regulators. Future studies of how ATM selects substrates to exert its multiple influences will significantly advance our understanding of the epigenetic programming underlying human neurodevelopment.

Materials and methods

Buffers

PBS: 137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4, 1.8 mM KH2PO4 (pH 7.4). PBST: PBS with 0.1% Triton X-100. HEPM: 25 mM HEPES (pH 6.9), 10 mM EGTA, 60 mM PIPES, 2 mM MgCl2. Immunofluorescence blocking solution: 1/3 Blocker Casein (Thermo Fisher Scientific), 2/3 HEPM with 0.05% TX-100. Buffer A: 10 mM HEPES (pH 7.9), 10 mM KCl, 1.5 mM MgCl2, 0.34 M sucrose, 10% glycerol. Buffer B: 3 mM EDTA, 0.2 mM EGTA. Buffer D: 400 mM KCl, 20 mM HEPES, 0.2 mM EDTA, 20% glycerol. ChIP lysis buffer 3: 10 mM Tris-HCl (pH 8.0), 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 0.1% sodium deoxycholate, 0.5% N-Lauroylsarcosine. ChIP wash buffer: 50 mM HEPES (pH 7.5), 500 mM LiCl, 1 mM EDTA, 1% NP-40, 0.7% Na-deoxycholate. ChIP elution buffer: 50 mM Tris-HCl (pH 8.0), 10 mM EDTA, 1% SDS. CUT&RUN Wash buffer: 20 mM HEPES (pH 7.5), 150 mM NaCl, 0.5 mM spermidine, protease inhibitor cocktail (Sigma-Aldrich 11873580001). CUT&RUN Binding buffer: 20 mM HEPES-KOH (pH 7.9), 10 mM KCl, 1 mM CaCl2, 1 mM MnCl2. CUT&RUN Digitonin buffer: CUT&RUN Wash buffer with 0.01% digitonin. CUT&RUN Antibody buffer: CUT&RUN Digitonin buffer with 2 mM EDTA. CUT&RUN 2X Stop buffer: 340 mM NaCl, 20 mM EDTA, 4 mM EGTA. CUT&RUN Stop buffer: Into 1 mL of 2X Stop buffer stock, add 5 μL of 10 mg/mL RNase A and 3.3 μL of 15 mg/mL. GlycoBlue Coprecipitant (Thermo Fisher AM9516)

Antibodies

S7 Table lists all antibodies and conditions used in this study.

ESC culture and mutagenesis

H9/WA09 (WiCell) hESCs were grown on Matrigel with reduced growth factors (Thermo Fisher Scientific, #35423) in mTeSR1 medium (STEMCELL Technologies, #85850) at 37°C and 5% CO2. The 53BP1 knock-in cell lines (53BP1 S25A 34–3, 34–4, 79–1, 79–3 and S25D 14–3, 14–15, 14–19, 17) and ATM KO cell lines (ATM-KO2, 3, 14, and 43) were generated using CRISPR/Cas9 gene-editing technology. Genome editing reagents were designed and validated in the Center for Advanced Genome Engineering at St. Jude Children’s Research Hospital. Briefly, a chemically modified sgRNA (Synthego) was precomplexed with SpCas9 protein (St. Jude Protein Production Core) and cotransfected with an ssODN donor template containing the desired modification into H9/WA09 cells via nucleofection (Amaxa P3 primary cell 4D nucleofector X kit L, Lonza) using the manufacturer’s recommended protocol. Transfected cells were sorted (BD FACSAria Fusion) onto Matrigel and allowed to grown single-cell clones. Clones were identified via targeted mi-seq using a 2-step PCR library setup as previously described [37]. Samples were demultiplexed using the index sequences, fastq files were generated, and NGS analysis was performed using CRIS.py [38]. S8 Table lists genome editing reagents and associated primers.

Neural progenitor cell generation and culture

ESCs were seeded onto AggreWell800 plates (STEMCELL Technologies, #34811) and fed with neural induction medium (STEMCELL Technologies, #05835) to form embryoid bodies. On day 5, embryoid bodies were replated onto Matrigel-treated 6-well plates in the same media. On day 17, cells were harvested as NPCs.

Nuclear extract preparation and western blotting

ESCs and NPCs were incubated in Buffer A + PI + DTT for 5 min on ice. After centrifugation at 1,750g for 2 min at 4°C, the nuclei pellet was washed in Buffer A and subsequently incubated for approximately 25 min in Buffer D + PI + DTT at 4°C with rotation to obtain the nuclear fraction. Nuclear extracts were separated by SDS–PAGE and transferred onto a nitrocellulose membrane (Bio-Rad). Membranes were blocked with 3% bovine serum albumin (BSA) in HEPM, incubated in primary antibodies (HEPM containing 1% BSA and 0.1% Triton X-100) overnight at 4°C, washed in PBS-T, incubated in IRDye-conjugated secondary antibodies (LI-COR), and imaged on an Odyssey Fc imaging system (LI-COR). Signals were quantitated with the Image Studio software (version 1.0.14; LI-COR).

Immunoprecipitation

Antibody was bound to protein A and protein G Dynabeads (Thermo Fisher 10002D and 10004D) for 2 h at room temperature. Nuclear extract was incubated with the Dynabeads-antibody complex for 5 h at 4°C, washed with PBST, and eluted with 0.1 M glycine (pH 2.3). Eluates were neutralized with 1/10 volume of 1.5 M Tris buffer (pH 8.8).

Cortical organoid differentiation

Cortical organoids were generated based on previously published methods with minor modifications [39,40]. In brief, hESC lines were expanded and dissociated to single cells using Accutase, seeded onto low-attachment V-bottom 96-well plates (Costar, #7007) at a density of 9,000 cells per well to aggregate into embryoid bodies. The embryoid bodies formation medium (DMEM/F-12 with 20% KO serum replacement, 3% ESC-quality FBS, 2 mM GlutaMAX, 0.1 mM nonessential amino acids) was supplemented with dorsomorphin (2 μM), WNT inhibitor (IWR1, 3 μM), TGF-β inhibitor (SB431542, 5 μM), and Rho kinase inhibitor (Y-27623, 20 μM). Starting from day 4, embryoid bodies were fed with cortical differentiation medium (Glasgow-MEM, 20% KSR, 0.1 mM NEAA, 1 mM sodium pyruvate, 0.1 mM β-ME, and 1% anti-anti), supplemented with WNT inhibitor (IWR1, 3 μM), TGF-β inhibitor (SB431542, 5 μM), cyclopamine (2.5 μM) and Rho kinase inhibitor (Y-27623, 20 μM). On day 17, embryoid bodies were embedded in Matrigel droplets and transferred onto low-attachment 6-wells and cultured in suspension using DMEM/F-12 supplemented with 1% N2 supplement, 1% lipid concentrate, 2% B27 supplement without vitamin A, and 1% anti-anti under 40% O2/5% CO2 conditions on shaker. Starting from day 30, medium was changed to 50% DMED/F-12, 50% neurobasal media, 0.5% N2 supplement, 1% GlutaMax, 0.05 mM NEAA, 0.025% human insulin, 0.1 mM β-ME, and 1% anti-anti, supplemented with 2% B27.

Immunofluorescence

Cells and cryosectioned organoids were blocked with IF blocking solution for 2 h at room temperature and primary antibodies (diluted in blocking buffer) added and incubated O/N at 4°C. After 3 washes in PBS-T, fluorescent dye-conjugated secondary antibodies (1:500, Alexa Fluor-CONJUGATED antibodies, Thermo Fisher Scientific) were added and incubated for 3 h at room temperature. Secondary was washed with PBS-T 3 times, and samples were washed and coverslips mounted with Prolong Glass Mounting Reagent (Thermo Fisher Scientific), which contains DAPI. Images were acquired with Zeiss LSM780.

Organoid feature characterization by image analysis

At days 35 and 55, bright-field images of organoids were captured with Axiocam 208 (Zeiss). Area of organoids, area of ventricular zone–like regions, and marker-positive cells were quantified by using the software FIJI: Signals-positive cells were identified based on signal and width thresholds. For ventricular zone–like region quantification, inner and outer edges of the regions in the image were manually traced, based on CTIP2-positive cells encircling the outer edges. FIJI was used to quantify area, perimeter, major and minor axes of the inner and outer traces. Mean perimeter and the difference between the major axes of the inner and outer traces were used to estimate the thickness of the structure. Mean Perimeter = (outer perimeter + inner perimeter) / 2. MajorAxisDiff = (outer major axis − inner major axis) / 2. MinorAxisDiff = (outer minor axis − inner minor axis) / 2. To quantify ZO-1-positive ventricular surfaces, ZO-1 signals were normalized by the Integral Image Filters plugin, and surface areas were manually traced for quantification. The VZ/SVZ structure was considered organized if PAX6-positive nuclei were densely packed with radial organization around ZO-1-positive ventricular surfaces. Ilastik [41] was used to quantify nuclear areas positive for different markers, using segmentation via a machine learning-based package and area quantification of segmented areas. Marker ratios were then calculated based on quantified areas.

Quantification of cell populations by fluorescence-activated cell sorting (FACS)

Nine to 12 organoids of each line were dissociated using the papain dissociation system (Worthington LK003153). Dissociated cells were fixed in 4% formaldehyde solution at 4°C overnight and washed once in 1X PBS. Then, cells were permeabilized in 1X PBST for 2 h at room temperature on an orbital shaker. Cells were blocked in IF blocking buffer (1/3 Blocker Casein (Thermo Fisher 37528), 2/3 HEPM with 0.05% Triton X-100) for 2 h at room temperature on a shaker. Primary antibodies in IF blocking buffer were mixed with cells at 4°C overnight followed by washing twice with 1X PBST. Secondary antibodies in IF blocking buffer were mixed with cells for 2 h at room temperature on a shaker. After washing cells once, a conjugated antibody was added and incubated for 2 h at room temperature on a shaker. Cells were washed one last time before resuspended in 1X PBS for FACS. FACSymphony A1 sorter was used for analysis. All the centrifugation steps were done at 500 × g for 4 min at room temperature. All washes were performed by incubating the cells with 1X PBS (after fixation) or PBST (after antibody staining) for 5 min at room temperature on a shaker. Primary antibodies used are Ki67 (Cell Signaling 9129), PAX6 (DSHB supernatant 1mL), CTIP2 (Abcam 18465), and cleaved Caspase3-AF405-conjugated (R&D Systems IC835V).

RNA-seq

Total RNA was extracted with TRIzol reagent (Invitrogen, #15596026) and Direct-zol RNA Microprep (Zymo Research, # R2062) by following manufacturer’s instructions. DNA digestion with DNase I was performed during RNA extraction. Paired-end 100-cycle sequencing was performed on NovaSeq6000 sequencer by following the manufacturer’s instructions (Illumina). Raw reads were first trimmed using TrimGalore (version 0.6.3) available at: https://www.bioinformatics.babraham.ac.uk/projects/trim_galore/, with parameters ‘—paired—retain_unpaired’. Filtered reads were then mapped to the Homo sapiens reference genome (GRCh38 + Gencode-v31) using STAR (version 2.7.9a) [42]. Gene-level read quantification was done using RSEM (version 1.3.1) [43]. To identify the differentially expressed genes between control and experimental samples, the variation in the library size between samples was first normalized by trimmed mean of M values (TMM) and genes with CPM < 1 in all samples were eliminated. Then, the normalized data were applied to linear modeling with the voom from the limma R package [44]. GSEA was performed against using the MSigDB database (version 7.1), and differentially expressed genes were ranked based on log2(FC) [45,46].

Protein extraction, digestion, and Tandem-Mass-Tag (TMT) labeling

Organoids were harvested on day 35, and the Matrigel droplets were eliminated by multiple ice-cold PBS washes. The organoid pellet was extracted in the lysis buffer (50 mM HEPES (pH 8.5), 8 M urea, and 0.5% sodium deoxycholate, 100 μl buffer per 10 mg tissue) with 1x PhosSTOP phosphatase inhibitor cocktail (Sigma-Aldrich). Protein concentration was estimated by a Coomassie stained short gel with BSA as a standard. About 600 μg each of protein samples was digested with LysC (Wako) at an enzyme-to-substrate ratio of 1:100 (w/w) for 2 h at room temperature in the presence of 1 mM DTT. The samples were then diluted to a final 2 M urea concentration with 50 mM HEPES (pH 8.5) and digested with Trypsin (Promega) at an enzyme-to-substrate ratio of 1:50 (w/w) for 3 h. The peptides were reduced by adding 1 mM DTT for 30 min at room temperature followed by alkylation with 10 mM iodoacetamide for 30 min in the dark at room temperature. The unreacted iodoacetamide was quenched with 30 mM DTT for 30 min. Finally, the digestion was terminated and acidified by adding trifluoroacetic acid to 1%, peptides desalted using Sep-Pak C18 cartridge (Waters), and dried by speed vac. The purified peptides were resuspended in 50 mM HEPES (pH 8.5) and labeled with 16-plex Tandem Mass Tag (TMTpro) reagents (Thermo Scientific) following the manufacturer’s recommendation. The TMT labeled samples were mixed equally, desalted using Sep-Pak C18 cartridge (Waters), and dried by speed vac.

Offline fractionation and two-dimensional liquid chromatography-tandem mass spectrometry (LC/LC-MS/MS)

The dried TMT mix was resuspended and fractionated on an offline HPLC (Agilent 1220) using basic pH reverse phase liquid chromatography (pH 8.0, XBridge C18 column, 4.6 mm × 25 cm, 3.5 μm particle size, Waters). A total of 160 one-minute fractions were collected and concatenated to 80 fractions. For whole proteome analysis, 10% of these 80 fractions was used. The remaining 90% of the 80 fractions were concatenated to 20 fractions for phophopeptide enrichment. Phosphopeptide enrichment was performed according to a previously published protocol [47]. The phosphopeptide enrichment eluents and the total proteome fractions were dried and resuspended in 5% formic acid and analyzed by acidic pH reverse phase LC-MS/MS analysis. The peptide samples were loaded on a nanoscale capillary reverse phase C18 column (New objective, 75 μm ID × approximately 15 cm, 1.9 μm C18 resin from Dr. Maisch GmbH) by a HPLC system (Thermo Ultimate 3000) and eluted by either a 125-min gradient (phosphofractions) or 110-min gradient for total proteome fractions. The eluted peptides were ionized by electrospray ionization and detected by an inline Orbitrap Fusion mass spectrometer (Thermo Scientific). For total proteome fractions, the mass spectrometer is operated in data-dependent mode with a survey scan in Orbitrap (60,000 resolution, 2 × 105 AGC target and 50 ms maximal ion time) and MS/MS high-resolution scans (60,000 resolution, 1 × 105 AGC target, 150 ms maximal ion time, 36.5 HCD normalized collision energy, 1 m/z isolation window, and 15-s dynamic exclusion). For phosphoproteome fractions, the mass spectrometer is operated in data-dependent mode with a survey scan in Orbitrap (60,000 resolution, 3 × 105 AGC target and 50 ms maximal ion time) and MS/MS high-resolution scans (60,000 resolution, 1 × 105 AGC target, 150 ms maximal ion time, 36.5 HCD normalized collision energy, 1 m/z isolation window, and 10-s dynamic exclusion).

Identification of proteins and phosphopeptides

The MS/MS raw data were processed by a tag-based hybrid search engine JUMP [48]. The data were searched against the UniProt human database (168,305 protein entries; downloaded in April 2020) concatenated with a reversed decoy database for evaluating FDR. Searches were performed using a 15-ppm mass tolerance for fragment ions, fully tryptic restriction with 2 maximal missed cleavages, 3 maximal modification sites, and the assignment of b and y ions. TMT tags on Lysine residues and N-termini (+304.2071453 Da) were used for static modifications and Met oxidation (+15.99492 Da) was considered as a dynamic modification. Phosphorylation (+79.96633 Da) was considered as a dynamic modification for STY residues. Putative peptide spectral matches (PSMs) were filtered by mass accuracy and then grouped by precursor ion charge state and filtered by JUMP-based matching scores (Jscore and ΔJn) to reduce FDR below 1% for proteins during the whole proteome analysis or 1% for phosphopeptides during the phosphoproteome analysis. Phosphosites were further evaluated by JUMPl program using the concept of the phosphoRS algorithm [49] to calculate phosphosite localization scores (Lscore, 0% to 100%) for each PSM.

Quantification of proteins and phosphopeptides

TMT reporter ion intensities of each PSM were extracted and corrected based on isotopic distribution of each labeling reagent. Those PSMs with very low intensities (e.g., minimum intensity of 1,000 and median intensity of 5,000) were excluded for quantification. Sample loading bias was mitigated by normalization with the trimmed median intensity of all PSMs. Protein or phosphopeptide relative intensities were calculated by dividing the intensity of each channel by the mean intensity. Protein or phosphopeptide absolute intensities were computed by multiplying the relative intensities by the grand-mean of 3 most highly abundant PSMs.

Differential expression analysis of proteins and phosphopeptides

Differentially expressed proteins between the 2 strains and 2 different doses were identified by the limma R package [50]. The Benjamini–Hochberg method was used to control multiple-testing correction, and proteins with an adjusted p-value of <0.05 and log2 fold change of >1.5 were defined as differentially expressed.

Pathway enrichment analysis for proteomics data

Pathway enrichment analysis was carried out to infer functional groups of proteins that were enriched in a given dataset. The 4 common pathway databases were used, including Gene Ontology (GO), KEGG, Hallmark, and Reactome. The analysis was performed using Fisher’s exact test with the Benjamini–Hochberg correction for multiple testing. A cutoff of adjusted p-value < 0.2 was used to identify significantly enriched pathways.

Estimation of kinase activity

Kinase activity was inferred based on known substrates in the PhosphoSitePlus database [51] using the IKAP algorithm [24]. The phosphoproteome data were normalized against the whole proteome. We performed 100 times of calculations to overcome the potential problem of local optimization.

Chromatin immunoprecipitation

Cells were harvested in PBS. Cytoplasmic fractions were extracted using buffer A with 1× protease inhibitors and 1 mM DTT. Nuclear pellets were cross-linked by 1.1% formaldehyde in buffer B with 1× protease inhibitors and 1 mM DTT; washed; and lysed in lysis buffer 3 with 1× protease inhibitors, 1 mM DTT, and 1 mM PMSF. The fixed and lysed nuclear extract was sonicated with Bioruptor Pico (Diagenode) 10 times for 15 s each, with 45-s intervals. Chromatin was added to Dynabeads (Life Technologies) prebound with 4 μg of antibodies for overnight incubation. After incubation, beads were washed and immunoprecipitates were eluted. DNA from eluates was recovered by the GeneJET FFPE DNA purification kit (Thermo Fisher Scientific, #K0882). DNA libraries were generated using the NEBNext Ultra DNA Library Prep kit (NEB, #E7370S) and sequenced at the St. Jude Hartwell Center.

CUT&RUN

Approximately 5 × 105 live cells were mixed with 5 × 104 Drosophila S2 cells per reaction. For CUT&RUN, we followed EpiCypher CUTANA protocol. In brief, we first isolated nuclei by incubating cells on ice for 5 min in Buffer A with protease inhibitor and 0.1% Triton X-100. After centrifugation at 1,750 × g for 2 min at 4°C, nuclei were resuspended in Wash buffer. Bio-Mag Plus Concanavalin-A (Con A) coated beads (Bangs Laboratories BP531) activated in Binding buffer were then added to the nuclei and rotated for 10 min at room temperature. About 1 μg primary antibody with 0.25 μg Spike-in antibody (Active Motif 61686) diluted in Antibody buffer was added to the bead-nuclei mixture and incubated for 2 h at room temperature. Beads were washed twice with Digitonin buffer and incubated with pAG-MNase for 10 min at room temperature. Beads were then washed twice with Digitonin buffer, incubated with 2 mM CaCl2 for 2 h at 4°C, and quenched by adding Stop buffer. DNA was released from the beads by incubating them for 10 min at 37°C and purified by CUTANA DNA purification kit (EpiCypher SKU:14–0050). Libraries were constructed using xGen ssDNA and Low-Input DNA Prep by following the manufacturer’s instructions (IDT 10009817) and sequenced at the St. Jude Hartwell Center.

Analysis of chromatin immunoprecipitation-sequencing and CUT&RUN

Approximately 50 bp single-end reads were obtained and aligned to human genome hg38 by BWA (version 0.7.170.7.12, default parameter). Duplicated reads were marked by the bamsormadup from the biobambam tool (version 2.0.87) available at https://www.sanger.ac.uk/tool/biobambam/. Uniquely mapped reads were kept by samtools (parameter “-q 1 -F 1804,” version 1.14). Fragments <2,000 bp were kept for peak calling, and bigwig files were generated for visualization. SICER [27] and macs2 [28] were both used for peak calling to identify both the narrow and broad peak correctly. With SICER, we assigned peaks that were at the top 1 percentile as the high-confidence peaks and the top 5 percentile as the low-confidence peaks. Two sets of peaks were generated: Strong peaks called with parameter “FDR < 0.05” by at least 1 method (macs2 or SICER) and weak peaks called with parameter “FDR < 0.5” by at least 1 method (macs2 or SICER). Peaks were considered reproducible if they were supported by 1 strong peaks and at least 1 weak peak in other replicates. For downstream analyses, heatmaps were generated by deepTools [52], and gene ontology was performed with Enrichr [53,54] and GSEA, in addition to custom R scripts. For differential peak analysis, peaks from 2 replicates were merged and counted for number of overlapping extended reads for each sample (bedtools v2.24.0) [55]. Then, we detected the differential peaks by the empirical Bayes method (eBayes function from the limma R package) [44]. For downstream analyses, heatmaps were generated by deepTools (v3.5.0) [56]. Peaks were annotated based on Gencode following this priority: “Promoter.Up”: if they fall within TSS– 2 kb, “Promoter.Down”: if they fall within TSS– 2 kb, “Exonic” or “intronic”: if they fall within an exon or intron of any isoform, “TES peaks”: if they fall within TES ± 2 kb, “distal5” or “distal3” if they are with 50 kb upstream of TSS or 50 kb downstream of TES, respectively, and they are classified as “intergenic” if they do not fit in any of the previous categories.

Supporting information

S1 Data. Numerical data used to generate summary data in this study.

(XLSX)

pbio.3002760.s001.xlsx (273.3KB, xlsx)
S1 Raw Images. Uncropped western blot images in this study.

(PDF)

pbio.3002760.s002.pdf (988.2KB, pdf)
S1 Fig. Characterization of 53BP1-pS25 and NPCs and genome editing of hESCs.

(A) Schematic diagram of neural differentiation of hESCs: neural induction, differentiation, and maturation media to form EBs, rosettes, NPCs, and neurons. (B) Principal component analysis of WT ESCs, NPCs, day 10 (D10) cortical organoids, and D17 cortical organoids. GSEA terms that are highly enriched in significantly (C) down-regulated and (D) up-regulated genes in WT NPCs compared to ESCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. (E) Immunofluorescence of NPC markers PAX6 and NESTIN. Bar, 50 μm. (F) Quantification of 53BP1-pS25-positive hESCs or hNPCs. Data are presented as the mean ± SEM, with p < 0.0001. (G) WB analysis of control cells and 53BP1-KO clones 415, 416, and 209, which are clones KO1, KO2, and KO3 in Yang and colleagues’ study [6]. (H) WB analysis of control and 53BP1-S25A hNPCs. The S25A mutation prohibits phosphorylation. (I) WB analysis of hESCs and hNPCs and quantification. (J) Schematic diagram of genome editing in hESCs. Guide RNA 6 were complexed with Cas9 proteins and used along single-stranded nucleotide donors to transfect hESCs. Individual clones from transfection were cultured, sequenced by mi-seq across the targeted 53BP1 locus, and established as >99% pure clonal lines. Diagram was generated using open-sourced images available at biorender.com. Underlying numerical values for figures are found in S1 Data. EB, embyoid body; ESC, embryonic stem cell; GSEA, gene set enrichment analysis; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WB, western blot; WT, wild type; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

(PDF)

pbio.3002760.s003.pdf (445.8KB, pdf)
S2 Fig. Generation and analyses of ATM-KO hESCs and cortical organoids.

(A) Alignment of WT and ATM-KO mutation sequences on 2 alleles (al) in the ATM locus. Red indicates the gRNA sequence. (B) WB analysis of WT and 4 ATM-KO hNPCs. (C) Principal component analysis showed the intermixing and similar RNA-seq profiles from hESCs of 7 WT, 4 53BP1-S25A, 4 53BP1-S25D, 4 ATM-KO, and 4 53BP1-KO lines. (D) Immunofluorescence showed similar expression of OCT4 and SSEA4 proteins in control and ATM-KO hESCs. Bar, 100 μm. (E) WB analysis of WT and 2 ATM-KO hNPCs. Quantification suggests reduction of γH2AX in ATM-KO hNPCs. Welch’s t test was used to perform pairwise comparisons of WT and ATM-KO. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; KO, knockout; WB, western blot; WT, wild type.

(PDF)

pbio.3002760.s004.pdf (385.8KB, pdf)
S3 Fig. Analysis of γH2AX and CC3 in cortical organoids.

(A) Immunofluorescence showed D35 ATM-KO and WT cortical organoids had similar γH2AX foci. Bar, 100 μm. FACS analysis of CC3 in (B) D21 and (C) D28 cortical organoids. Two biological replicates were done, and each data point was based on 3 technical replicate analyses of 10–12 cortical organoids. (D, E) Immunofluorescence and quantification of CC3 in D28 cortical organoids. Bar, 100 μm. Graphs are presented in ratios (out of 1), with **, p < 0.01; ****, p < 0.0001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; CC3, cleaved-caspase 3; KO, knockout; WT, wild type.

(PDF)

pbio.3002760.s005.pdf (207.8KB, pdf)
S4 Fig. Analysis of cell proliferation in cortical organoids.

FACS analysis of PAX6 and KI67 in (A, B) D28 and (C) D35 cortical organoids. Each data point was based on the 3 technical replicate analyses of 10 cortical organoids. (D) Quantification of KI67/PAX6 ratios in immunofluorescence of D35 cortical organoids. Each data point represents quantification of cells in 1 cortical organoid. (E, F) Immunofluorescence and quantification of H3-pS10 (PH3) in D28 cortical organoids. Bar, 100 μm. (F-H) Immunofluorescence and quantification of PH3 and KI67 in D35 cortical organoids. Bar, 100 μm. ***, p < 0.001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data.

(PDF)

pbio.3002760.s006.pdf (338.2KB, pdf)
S5 Fig. Immunofluorescence analyses of cortical organoids and NPCs.

Immunofluorescence of (A) NEUN and (E) ZO-1 and PAX6 in D37 cortical organoids. Bar, 100 μm. (B) Immunofluorescence of ZO-1 in D28 cortical organoids. Bar, 100 μm. Quantification of the (C) number and (D) surface area of ZO-1-positive ventricles in D28 cortical organoids. *, p < 0.05; ***, p <0 .001; ns, not significant by two-way ANOVA test. (F) Bright-field images of cortical organoids formed by ATM-KO2, 3, 14, 43, and WT control at day 55 of differentiation. Bar, 1.5 mm. (G) The size of cortical organoids was compared between groups by one-way ANOVA with Dunnett’s multiple comparisons test, with ns, not significant and ***, p < 0.001. n = 13 organoids/group. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; NPC, neural progenitor cell; WT, wild type.

(PDF)

pbio.3002760.s007.pdf (389.3KB, pdf)
S6 Fig. Characterization of NPCs and D35 cortical organoids.

(A) Immunofluorescence of PAX6 and NES in NPCs. Bar, 50 μm. (B) Principal component analysis of proteomics data of D35 WT and ATM-KO cortical organoids. GSEA terms that are highly enriched in significantly (C) higher and (D) lower total proteins in D35 ATM-KO versus WT cortical organoids. (E) GSEA terms that are highly enriched in significantly higher phosphoproteins, which were normalized to total proteomics, in D35 ATM-KO versus WT cortical organoids. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type.

(PDF)

pbio.3002760.s008.pdf (309.8KB, pdf)
S7 Fig. Kinase activities in cortical organoids and characterization of the 53BP1-S25A and 53BP1-S25D hESCs.

Heatmaps showing relative phosphorylation levels of (A) 7 MAPK9 substrates that are significantly lower and (B) 7 CDK5 substrates that are significantly higher in D35 ATM-KO versus WT cortical organoids. (C) Heatmaps showing activity of selected protein kinases between ATM-KO3, ATM-KO4, and WT cell lines. (D) Alignment of WT and 53BP1-S25A and S25D mutation sequences on 2 alleles (al). Red indicates the gRNA sequence. Underline indicates codon encoding the WT serine 25, mutant alanine, or mutant aspartic acid. (E) WB analysis of control and 53BP1-S25D hNPCs, which have comparable levels of 53BP1 protein. (F) Transcripts per million values of 10 pluripotency genes were used for comparison to show that control, 53BP1-S25A, and 53BP1-S25D hESCs did not differ in pluripotency. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; KO, knockout; WB, western blot; WT, wild type; 53BP1, p53 binding protein 1.

(PDF)

pbio.3002760.s009.pdf (393.5KB, pdf)
S8 Fig. Characterization of the 53BP1-S25A and 53BP1-S25D hESCs and cortical organoids.

(A) Immunofluorescence showed similar expression of OCT4 and SSEA4 proteins in WT, 53BP1-S25A, and 53BP1-S25D hESCs. Bar, 100 μm. Immunofluorescence of (B) KI67 and (D) PH3 in cryosections of cortical organoids at day 35 of differentiation. Bar, 100 μm. (C) Quantification of KI67-positive cells in D35 cortical organoids. Data points represent single organoids. The mean ± SEM values were compared by one-way ANOVA with Dunnett’s multiple comparisons test to yield ****, ***, and ** indicating p < 0.0001, 0.001, and 0.01, respectively. n = 3 organoids/group. Underlying numerical values for figures are found in S1 Data.

(PDF)

pbio.3002760.s010.pdf (402.8KB, pdf)
S9 Fig. Analysis of γH2AX and CC3 in cortical organoids.

Immunofluorescence of (A) γH2AX in D35 cortical organoids and (D) CC3 in D28 cortical organoids. Bar, 100 μm. CC3 quantification by FACS of (B) D21 and (C) D28 cortical organoids. For each datapoint, 10–12 organoids from each line were analyzed via 3 technical replicates, and data from 4 mutant lines were consolidated to achieve rigorous comparisons. **, p < 0.01 and ns, not significant by two-way ANOVA test. (E) CC3 quantification of immunofluorescence images of D28 cortical organoids. For each line, 4–6 images and >10,000 cells were analyzed. *, p < 0.05; **, p < 0.01; ns, not significant by two-way ANOVA test. Graphs in (B, C, E) are presented in ratios (out of 1). Underlying numerical values for figures are found in S1 Data.

(PDF)

pbio.3002760.s011.pdf (333.1KB, pdf)
S10 Fig. 53BP1-pS25 promotes ventricle formation in cortical organoids.

Quantification of the (A) number and (B) surface area of ZO-1-positive ventricles. (C) Immunofluorescence of ZO-1 in D28 cortical organoids. Bar, 100 μm. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data.

(PDF)

pbio.3002760.s012.pdf (268.4KB, pdf)
S11 Fig. Characterization of 53BP1-S25A and 53BP1-S25D cortical organoids.

(A) Bright-field images of cortical organoids formed by cell lines 53BP1-S25A 34–3, 34–4, 79–1, 79–3 and S25D 14–3, 14–15, 14–19, 17, and 2 WT control at day 55 of differentiation. Bar, 1.5 mm. Blue transparent structures around organoids are Matrigel embedment. (B) At day 55 of differentiation, the size of cortical organoids was compared between groups. (C) The growth (comparing organoids at days 35 and 55) of cortical organoids were compared between groups. Data points represent single organoids. The mean ± SEM values were compared by one-way ANOVA with Dunnett’s multiple comparisons test to yield **** and ** indicating p < 0.0001 and 0.01, respectively. n = 15–36 organoids/group. (D) Two genes overlapped between up-regulated genes in 53BP1-S25A versus WT and down-regulated genes in 53BP1-S25D versus WT cortical organoids. No gene overlapped between down-regulated genes in 53BP1-S25A versus WT and up-regulated genes in 53BP1-S25D versus WT cortical organoids. (E) Down-regulated GSEA terms between 53BP1-S25A versus WT and 53BP1-S25D versus WT were not highly overlapped. Ten GSEA terms were specific to 53BP1-S25D versus WT. Underlying numerical values for figures are found in S1 Data.

(PDF)

pbio.3002760.s013.pdf (457.1KB, pdf)
S12 Fig. Characterization of NPCs and comparative analyses of RNA-seq data.

(A) Immunofluorescence of NPC markers PAX6 and NESTIN. Bar, 50 μm. GSEA identified top enrichment of differentially expressed genes in (B, D) 53BP1-S25A or (C, E) S25D versus WT NPCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. Venn diagrams depict overlaps between down-regulated genes in ATM-KO with 53BP1- (F) S25A or (G) S25D cortical organoids. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type.

(PDF)

pbio.3002760.s014.pdf (413.2KB, pdf)
S13 Fig. Comparisons of RNA-seq data and 53BP1 ChIP-seq analyses.

(A) GSEA graphs showed that up-regulated genes in 53BP1-S25A or S25D vs. WT had significant enrichment in down-regulated genes of ATM-KO vs. WT cortical organoids. P values were calculated by the hypergeometric test, assuming normal data distribution. (B) Concordantly differential expression of genes in 53BP1-S25D vs. WT were enriched in those in 53BP1-S25A vs. WT. (C) Concordantly differential expression of genes in 53BP1-S25A vs. WT were enriched in those in 53BP1-S25D vs. WT. For (A-C), P values were calculated by the hypergeometric test, assuming normal data distribution. (D) Proportions of 53BP1 binding to genomic features. 53BP1 ChIP-seq tracks at loci of representative (E) up-regulated and (F) down-regulated genes in 53BP1-S25A and S25D versus WT D35 cortical organoids. (G) S25A and S25D down-regulate 53BP1 targets that are enriched in IRE1-mediated unfolded protein response, regulation of cellular response to stress, iron import into cells, and regulation of apoptosis. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; WT, wild type.

(PDF)

pbio.3002760.s015.pdf (530.4KB, pdf)
S14 Fig. 53BP1 ChIP-seq and 53BP1-pS25 CUT&RUN.

(A) MA plot displays 53BP1 ChIP-seq signals at genomic sites that are significantly different in ATM-KO vs. WT NPCs. Proportions of genomic features and gene ontology of genes with (B) higher or (C) lower 53BP1 binding in ATM-KO vs. WT NPCs. (D) Proportions of 53BP1-pS25 binding to genomic features. (E) GSEA identified top enrichment of genes occupied by 53BP1-pS25 in WT NPCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type.

(PDF)

pbio.3002760.s016.pdf (222KB, pdf)
S15 Fig. Differential 53BP1 ChIP-seq in 53BP1-WT, S25A, and S25D NPCs.

(A) Principal component analysis of top 3,000 most variable peaks in 53BP1 ChIP-seq of 53BP1-WT, S25A, and S25D NPCs. Two independent cell lines for each group were used for ChIP-seq. Proportions of genomic features in regions with significantly different 53BP1 ChIP-seq in (B) 53BP1-S25A vs. WT and (C) 53BP1-S25D vs. WT, using the criterion of FC>2 and p < 0.05. (D) Heatmaps aligning peaks with significantly different 53BP1 ChIP-seq in 53BP1-S25A vs. S25D. Control regions are those, after voom normalization, showed the least changes and served as semi-independent validation of differential ChIP-seq analysis. Bubble graphs present top enriched categories of genes that had significantly lower 53BP1 ChIP-seq in (E) 53BP1-S25A vs. WT and (F) 53BP1-S25D vs. WT. Underlying numerical values for figures are found in S1 Data. FC, fold-change; NPC, neural progenitor cell; WT, wild type.

(PDF)

pbio.3002760.s017.pdf (297.8KB, pdf)
S16 Fig. Analysis of ATM activities during the inhibition of TGFβ, WNT, and HH signaling.

(A) Schematic diagram of neural specification of hESCs with HH (SB421542), TGFβ (dorsomorphin), and WNT (IWR1e and cyclopamine) signaling inhibitors. Nuclear extract was harvested on day 4 and day 10. (B) WB analysis of day 4 samples. (C) Quantification of day 4 WB. Data are presented as the mean ± SEM, and Student t test was performed for pairwise comparisons. n.s., *, and ** indicate not significant, p < 0.05, and p < 0.01, respectively. (D) WB analysis of day 10 samples. (E) Quantification of day 10 WB. Data are presented as the mean ± SEM, and Student t test was performed for pairwise comparisons. n.s., *, and ** indicate not significant, p < 0.05, and p < 0.01, respectively. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; hESC, human embryonic stem cell; WB, western blot.

(PDF)

pbio.3002760.s018.pdf (281.7KB, pdf)
S17 Fig. RNF168-KO alters key genetic programs and 53BP1-pS25 binding to chromatin.

(A) Alignment of WT and RNF168-KO mutation sequences in the RNF168 locus. Red indicates the gRNA sequences. (B) WB analysis of WT and RNF168-KO hESCs. (C) RT-qPCR analysis showing that pluripotent genes in RNF168-KO were expressed higher or the same as those in WT. RNF168-KO did not reduce pluripotent gene expression. *, p < 0.05; ns, not significant by two-way ANOVA text. (D) Immunofluorescence showed similar expression of OCT4 and SSEA4 proteins in WT and RNF168-KO hESCs. Bar, 100 μm. (E) Immunofluorescence of showed similar expression of PAX6 and NES in NPCs. WT and RNF168-KO NPCs. Bar, 50 μm. Functional terms that are highly enriched in (F) up-regulated and (G) down-regulated genes in RNF168-KO D35 cortical organoids. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. (H) Heatmaps aligning peaks with 53BP1-pS25 CUT&RUN signals that were gained, the same, or lost in RNF168-KO vs. WT NPCs, using the criterion of FC>2 and p < 0.05. n = numbers of peaks. Regions with the same signals, are n = 899, which showed the least changes after voom normalization and served as semi-independent validation of differential ChIP-seq analysis. (I) Functional terms of 53BP1-pS25-bound genes in WT NPCs. % Match, % of genes in the enriched term that overlap the differentially bound genes. (J) Number of differentially expressed genes identified by comparison of RNF168-KO vs. WT NPCs at p < 0.05. Of these genes, we list the numbers of 53BP1-pS25-bound targets and targets with higher or lower 53BP1-pS25 CUT&RUN signals in RNF168-KO NPCs. Underlying numerical values for figures are found in S1 Data. FC, fold-change; hESC, human embryonic stem cell; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; RT-qPCR, quantitative reverse transcription PCR; WB, western blot; WT, wild type; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

(PDF)

pbio.3002760.s019.pdf (568.5KB, pdf)
S1 Table. All cell lines generated for this study were treated with trypsin and Wright’s stain and then analyzed by the Cytogenetic Shared Resource at St. Jude.

Typically normal karyotypes and 3 abnormalities are shown.

(PDF)

pbio.3002760.s020.pdf (1.1MB, pdf)
S2 Table. The expression of forebrain, midbrain, and hindbrain markers in D35 WT and ATM-KO cortical organoids.

Data suggest that D35 ATM-KO cortical organoids specified to the forebrain lineage.

(PDF)

pbio.3002760.s021.pdf (37.2KB, pdf)
S3 Table. List of phosphoproteins, normalized to total protein levels, that were significantly lower in D35 ATM-KO versus WT cortical organoids.

(XLSX)

pbio.3002760.s022.xlsx (22.9KB, xlsx)
S4 Table. Normalized (to total proteome) levels of phosphopeptide substrates of MAPK9 and CDK5 in D35 WT and ATM-KO cortical organoids.

(XLSX)

pbio.3002760.s023.xlsx (172.7KB, xlsx)
S5 Table. Two-sample t test examines the sizes of cortical organoids that change between day 35 and day 55 of differentiation.

Data from WT and 53BP1 mutants are compared pairwise by using the two-sample t test. The sizes of organoids are significantly different between each comparison pair (all p < 0.05).

(PDF)

pbio.3002760.s024.pdf (163.4KB, pdf)
S6 Table. The changes in organoid size at days 35 and 55 of differentiation were compared to yield S3C Fig.

This table lists the calculation for different combinations of data and the descriptive statistics.

(PDF)

pbio.3002760.s025.pdf (150.4KB, pdf)
S7 Table. Primary antibodies used in this study.

(PDF)

pbio.3002760.s026.pdf (21.1KB, pdf)
S8 Table. gRNA sequences used in this study.

(PDF)

pbio.3002760.s027.pdf (14.3KB, pdf)

Acknowledgments

The authors thank A. Andersen and I. Chen for discussions and editing the manuscript; A. N. Kettenbach for advice; J. Houston and K. Lowe for FACS; P. Sinojia and E. Rivera-Peraza for preliminary experiments and data analyses. Sequencing was performed at the Harwell Center for Biotechnology, images were acquired at the Cell & Tissue Imaging Center, and karyotyping was analyzed by J. Wilbourne and V. Valentine at the Cytogenetics Core; all are supported by SJCRH and NCI P30 (CA021765).

Abbreviations

ATM

ataxia telangiectasia mutated

BSA

bovine serum albumin

FDR

false discovery rate

GO

Gene Ontology

GSEA

gene set enrichment analysis

hESC

human embryonic stem cell

KO

knockout

NPC

neural progenitor cell

PSM

peptide spectral match

TMT LC-MS/MS

liquid chromatography-tandem mass spectrometry

WB

western blot

WT

wild type

53BP1

p53 binding protein 1

53BP1-pS25

53BP1 phosphorylated at serine 25

Data Availability

All sequencing data are deposited in NCBI GEO database under accession number GSE231321. Codes for analyzing sequencing data are deposited in https://doi.org/10.6084/m9.figshare.7411835. Mass spectrometry data were deposited in ProteomXchange, with project accession number PXD041699. Numerical data are in S1 Data, and uncropped Western blot images are in S1 Raw Images.

Funding Statement

This work was supported by the American Lebanese Syrian Associated Charities (https://www.stjude.org/ to JCP), American Cancer Society (https://www.cancer.org/; 132096-RSG-18-032-01-DDC to JCP), and NIH (https://www.nih.gov/; 1R01GM134358-05 to JCP). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Hanawalt PC, Spivak G. Transcription-coupled DNA repair: two decades of progress and surprises. Nat Rev Mol Cell Biol. 2008;9(12):958–970. doi: 10.1038/nrm2549 . [DOI] [PubMed] [Google Scholar]
  • 2.Mellon I, Spivak G, Hanawalt PC. Selective removal of transcription-blocking DNA damage from the transcribed strand of the mammalian DHFR gene. Cell. 1987;51(2):241–249. doi: 10.1016/0092-8674(87)90151-6 . [DOI] [PubMed] [Google Scholar]
  • 3.Nelson P. Transport of torsional stress in DNA. Proc Natl Acad Sci U S A. 1999;96(25):14342–14347. doi: 10.1073/pnas.96.25.14342 ; PubMed Central PMCID: PMC24438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Panier S, Boulton SJ. Double-strand break repair: 53BP1 comes into focus. Nat Rev Mol Cell Biol. 2014;15(1):7–18. doi: 10.1038/nrm3719 . [DOI] [PubMed] [Google Scholar]
  • 5.Cuella-Martin R, Oliveira C, Lockstone HE, Snellenberg S, Grolmusova N, Chapman JR. 53BP1 Integrates DNA Repair and p53-Dependent Cell Fate Decisions via Distinct Mechanisms. Mol Cell. 2016;64(1):51–64. doi: 10.1016/j.molcel.2016.08.002 ; PubMed Central PMCID: PMC5065530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yang X, Xu B, Mulvey B, Evans M, Jordan S, Wang YD, et al. Differentiation of human pluripotent stem cells into neurons or cortical organoids requires transcriptional co-regulation by UTX and 53BP1. Nat Neurosci. 2019;22(3):362–373. Epub 20190204. doi: 10.1038/s41593-018-0328-5 ; PubMed Central PMCID: PMC6511450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kleiner RE, Verma P, Molloy KR, Chait BT, Kapoor TM. Chemical proteomics reveals a gammaH2AX-53BP1 interaction in the DNA damage response. Nat Chem Biol. 2015;11(10):807–814. Epub 20150907. doi: 10.1038/nchembio.1908 ; PubMed Central PMCID: PMC4589150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wilson MD, Benlekbir S, Fradet-Turcotte A, Sherker A, Julien JP, McEwan A, et al. The structural basis of modified nucleosome recognition by 53BP1. Nature. 2016;536(7614):100–103. Epub 20160727. doi: 10.1038/nature18951 . [DOI] [PubMed] [Google Scholar]
  • 9.Fradet-Turcotte A, Canny MD, Escribano-Diaz C, Orthwein A, Leung CC, Huang H, et al. 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature. 2013;499(7456):50–54. Epub 20130612. doi: 10.1038/nature12318 ; PubMed Central PMCID: PMC3955401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ward IM, Minn K, Jorda KG, Chen J. Accumulation of checkpoint protein 53BP1 at DNA breaks involves its binding to phosphorylated histone H2AX. J Biol Chem. 2003;278(22):19579–19582. Epub 20030415. doi: 10.1074/jbc.C300117200 . [DOI] [PubMed] [Google Scholar]
  • 11.Chapman JR, Barral P, Vannier JB, Borel V, Steger M, Tomas-Loba A, et al. RIF1 Is Essential for 53BP1-Dependent Nonhomologous End Joining and Suppression of DNA Double-Strand Break Resection. Mol Cell. 2021;81(13):2868. doi: 10.1016/j.molcel.2021.06.015 ; PubMed Central PMCID: PMC8260204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Isobe SY, Nagao K, Nozaki N, Kimura H, Obuse C. Inhibition of RIF1 by SCAI Allows BRCA1- Mediated Repair. Cell Rep. 2017;20(2):297–307. doi: 10.1016/j.celrep.2017.06.056 . [DOI] [PubMed] [Google Scholar]
  • 13.Dobersch S, Rubio K, Singh I, Gunther S, Graumann J, Cordero J, et al. Positioning of nucleosomes containing gamma-H2AX precedes active DNA demethylation and transcription initiation. Nat Commun. 2021;12(1):1072. Epub 20210216. doi: 10.1038/s41467-021-21227-y ; PubMed Central PMCID: PMC7886895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bunch H, Lawney BP, Lin YF, Asaithamby A, Murshid A, Wang YE, et al. Transcriptional elongation requires DNA break-induced signalling. Nat Commun. 2015;6:10191. Epub 2015/12/17. doi: 10.1038/ncomms10191 ; PubMed Central PMCID: PMC4703865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Harding SM, Coackley C, Bristow RG. ATM-dependent phosphorylation of 53BP1 in response to genomic stress in oxic and hypoxic cells. Radiother Oncol. 2011;99(3):307–312. Epub 20110615. doi: 10.1016/j.radonc.2011.05.039 . [DOI] [PubMed] [Google Scholar]
  • 16.Jowsey P, Morrice NA, Hastie CJ, McLauchlan H, Toth R, Rouse J. Characterisation of the sites of DNA damage-induced 53BP1 phosphorylation catalysed by ATM and ATR. DNA Repair (Amst). 2007;6(10):1536–1544. Epub 20070605. doi: 10.1016/j.dnarep.2007.04.011 . [DOI] [PubMed] [Google Scholar]
  • 17.Isono M, Niimi A, Oike T, Hagiwara Y, Sato H, Sekine R, et al. BRCA1 Directs the Repair Pathway to Homologous Recombination by Promoting 53BP1 Dephosphorylation. Cell Rep. 2017;18(2):520–532. doi: 10.1016/j.celrep.2016.12.042 . [DOI] [PubMed] [Google Scholar]
  • 18.Munoz IM, Jowsey PA, Toth R, Rouse J. Phospho-epitope binding by the BRCT domains of hPTIP controls multiple aspects of the cellular response to DNA damage. Nucleic Acids Res. 2007;35(16):5312–5322. Epub 20070808. doi: 10.1093/nar/gkm493 ; PubMed Central PMCID: PMC2018624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gupta A, Hunt CR, Hegde ML, Chakraborty S, Chakraborty S, Udayakumar D, et al. MOF phosphorylation by ATM regulates 53BP1-mediated double-strand break repair pathway choice. Cell Rep. 2014;8(1):177–189. Epub 20140619. doi: 10.1016/j.celrep.2014.05.044 ; PubMed Central PMCID: PMC4300955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wang D, Ma J, Botuyan MV, Cui G, Yan Y, Ding D, et al. ATM-phosphorylated SPOP contributes to 53BP1 exclusion from chromatin during DNA replication. Sci Adv. 2021;7(25). Epub 20210618. doi: 10.1126/sciadv.abd9208 ; PubMed Central PMCID: PMC8213225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Carlessi L, De Filippis L, Lecis D, Vescovi A, Delia D. DNA-damage response, survival and differentiation in vitro of a human neural stem cell line in relation to ATM expression. Cell Death Differ. 2009;16(6):795–806. Epub 20090220. doi: 10.1038/cdd.2009.10 . [DOI] [PubMed] [Google Scholar]
  • 22.Yang DQ, Kastan MB. Participation of ATM in insulin signalling through phosphorylation of eIF-4E-binding protein 1. Nat Cell Biol. 2000;2(12):893–898. doi: 10.1038/35046542 . [DOI] [PubMed] [Google Scholar]
  • 23.Kastan MB, Lim DS. The many substrates and functions of ATM. Nat Rev Mol Cell Biol. 2000;1(3):179–186. doi: 10.1038/35043058 . [DOI] [PubMed] [Google Scholar]
  • 24.Mischnik M, Sacco F, Cox J, Schneider HC, Schafer M, Hendlich M, et al. IKAP: A heuristic framework for inference of kinase activities from Phosphoproteomics data. Bioinformatics. 2016;32(3):424–431. Epub 20151201. doi: 10.1093/bioinformatics/btv699 . [DOI] [PubMed] [Google Scholar]
  • 25.Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER 3rd, Hurov KE, Luo J, et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 2007;316(5828):1160–1166. doi: 10.1126/science.1140321 . [DOI] [PubMed] [Google Scholar]
  • 26.Chen Z, Cole PA. Synthetic approaches to protein phosphorylation. Curr Opin Chem Biol. 2015;28:115–122. Epub 20150718. doi: 10.1016/j.cbpa.2015.07.001 ; PubMed Central PMCID: PMC4624483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Xu S, Grullon S, Ge K, Peng W. Spatial clustering for identification of ChIP-enriched regions (SICER) to map regions of histone methylation patterns in embryonic stem cells. Methods Mol Biol. 2014;1150:97–111. Epub 2014/04/20. doi: 10.1007/978-1-4939-0512-6_5 ; PubMed Central PMCID: PMC4152844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008;9(9):R137. Epub 2008/09/19. doi: 10.1186/gb-2008-9-9-r137 ; PubMed Central PMCID: PMC2592715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448(7153):553–560. Epub 20070701. doi: 10.1038/nature06008 ; PubMed Central PMCID: PMC2921165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Doil C, Mailand N, Bekker-Jensen S, Menard P, Larsen DH, Pepperkok R, et al. RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins. Cell. 2009;136(3):435–446. doi: 10.1016/j.cell.2008.12.041 . [DOI] [PubMed] [Google Scholar]
  • 31.Paull TT. Mechanisms of ATM Activation. Annu Rev Biochem. 2015;84:711–738. Epub 20150112. doi: 10.1146/annurev-biochem-060614-034335 . [DOI] [PubMed] [Google Scholar]
  • 32.Le Belle JE, Orozco NM, Paucar AA, Saxe JP, Mottahedeh J, Pyle AD, et al. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner. Cell Stem Cell. 2011;8(1):59–71. doi: 10.1016/j.stem.2010.11.028 ; PubMed Central PMCID: PMC3018289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Shreeram S, Demidov ON, Hee WK, Yamaguchi H, Onishi N, Kek C, et al. Wip1 phosphatase modulates ATM-dependent signaling pathways. Mol Cell. 2006;23(5):757–764. doi: 10.1016/j.molcel.2006.07.010 . [DOI] [PubMed] [Google Scholar]
  • 34.Chun HH, Gatti RA. Ataxia-telangiectasia, an evolving phenotype. DNA Repair (Amst). 2004;3(8–9):1187–1196. doi: 10.1016/j.dnarep.2004.04.010 . [DOI] [PubMed] [Google Scholar]
  • 35.Lavin MF. Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signalling and cancer. Nat Rev Mol Cell Biol. 2008;9(10):759–769. doi: 10.1038/nrm2514 . [DOI] [PubMed] [Google Scholar]
  • 36.McKinnon PJ. ATM and the molecular pathogenesis of ataxia telangiectasia. Annu Rev Pathol. 2012;7:303–321. Epub 20111024. doi: 10.1146/annurev-pathol-011811-132509 . [DOI] [PubMed] [Google Scholar]
  • 37.Sentmanat MF, Peters ST, Florian CP, Connelly JP, Pruett-Miller SM. A Survey of Validation Strategies for CRISPR-Cas9 Editing. Sci Rep. 2018;8(1):888. doi: 10.1038/s41598-018-19441-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Connelly JP, Pruett-Miller SM. CRIS.py: A Versatile and High-throughput Analysis Program for CRISPR-based Genome Editing. Sci Rep. 2019;9():4194. Epub 20190312. doi: 10.1038/s41598-019-40896-w ; PubMed Central PMCID: PMC6414496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kadoshima T, Sakaguchi H, Nakano T, Soen M, Ando S, Eiraku M, et al. Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell-derived neocortex. Proc Natl Acad Sci U S A. 2013;110(50):20284–20289. Epub 2013/11/28. doi: 10.1073/pnas.1315710110 ; PubMed Central PMCID: PMC3864329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lancaster MA, Knoblich JA. Generation of cerebral organoids from human pluripotent stem cells. Nat Protoc. 2014;9(10):2329–2340. doi: 10.1038/nprot.2014.158 ; PubMed Central PMCID: PMC4160653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Berg S, Kutra D, Kroeger T, Straehle CN, Kausler BX, Haubold C, et al. ilastik: interactive machine learning for (bio)image analysis. Nat Methods. 2019;16(12):1226–1232. Epub 20190930. doi: 10.1038/s41592-019-0582-9 . [DOI] [PubMed] [Google Scholar]
  • 42.Dobin A, Gingeras TR. Mapping RNA-seq Reads with STAR. Curr Protoc Bioinformatics. 2015;51:11 4 1–4 9. Epub 2015/09/04. doi: 10.1002/0471250953.bi1114s51 ; PubMed Central PMCID: PMC4631051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323. Epub 2011/08/06. doi: 10.1186/1471-2105-12-323 ; PubMed Central PMCID: PMC3163565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Law CW, Chen Y, Shi W, Smyth GK. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15(2):R29. doi: 10.1186/gb-2014-15-2-r29 ; PubMed Central PMCID: PMC4053721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1(6):417–425. Epub 2016/01/16. doi: 10.1016/j.cels.2015.12.004 ; PubMed Central PMCID: PMC4707969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. Epub 2005/10/04. doi: 10.1073/pnas.0506580102 ; PubMed Central PMCID: PMC1239896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tan H, Wu Z, Wang H, Bai B, Li Y, Wang X, et al. Refined phosphopeptide enrichment by phosphate additive and the analysis of human brain phosphoproteome. Proteomics. 2015;15(2–3):500–507. Epub 20141215. doi: 10.1002/pmic.201400171 ; PubMed Central PMCID: PMC4598062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang X, Li Y, Wu Z, Wang H, Tan H, Peng J. JUMP: a tag-based database search tool for peptide identification with high sensitivity and accuracy. Mol Cell Proteomics. 2014;13(12):3663–3673. Epub 2014/09/10. doi: 10.1074/mcp.O114.039586 ; PubMed Central PMCID: PMC4256513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Taus T, Kocher T, Pichler P, Paschke C, Schmidt A, Henrich C, et al. Universal and confident phosphorylation site localization using phosphoRS. J Proteome Res. 2011;10(12):5354–5362. Epub 20111110. doi: 10.1021/pr200611n . [DOI] [PubMed] [Google Scholar]
  • 50.Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47. Epub 2015/01/22. doi: 10.1093/nar/gkv007 ; PubMed Central PMCID: PMC4402510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hornbeck PV, Kornhauser JM, Latham V, Murray B, Nandhikonda V, Nord A, et al. 15 years of PhosphoSitePlus(R): integrating post-translationally modified sites, disease variants and isoforms. Nucleic Acids Res. 2019;47(D1):D433–D441. doi: 10.1093/nar/gky1159 ; PubMed Central PMCID: PMC6324072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ramirez F, Dundar F, Diehl S, Gruning BA, Manke T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 2014;42(Web Server issue):W187–W191. Epub 2014/05/07. doi: 10.1093/nar/gku365 ; PubMed Central PMCID: PMC4086134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90–W97. Epub 2016/05/05. doi: 10.1093/nar/gkw377 ; PubMed Central PMCID: PMC4987924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128. Epub 2013/04/17. doi: 10.1186/1471-2105-14-128 ; PubMed Central PMCID: PMC3637064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26(6):841–842. Epub 2010/01/30. doi: 10.1093/bioinformatics/btq033 ; PubMed Central PMCID: PMC2832824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ramirez F, Ryan DP, Gruning B, Bhardwaj V, Kilpert F, Richter AS, et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016;44(W1):W160–W165. Epub 2016/04/16. doi: 10.1093/nar/gkw257 ; PubMed Central PMCID: PMC4987876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.UTX and MLL4 Coordinately Regulate Transcriptional Programs for Cell Proliferation and Invasiveness in Breast Cancer Cells. Cancer Res. 2017;77(9):2553. doi: 10.1158/0008-5472.CAN-17-0639 . [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Lucas Smith

4 Sep 2023

Dear Dr Peng,

Thank you for submitting your manuscript entitled "Phosphorylation of 53BP1 by ATM controls neurodevelopmental programsin cortical brain organoids" for consideration as a Research Article by PLOS Biology.

Your manuscript has now been evaluated by the PLOS Biology editorial staff as well as by an academic editor with relevant expertise and I am writing to let you know that we would like to send your submission out for external peer review.

However, before we can send your manuscript to reviewers, we need you to complete your submission by providing the metadata that is required for full assessment. To this end, please login to Editorial Manager where you will find the paper in the 'Submissions Needing Revisions' folder on your homepage. Please click 'Revise Submission' from the Action Links and complete all additional questions in the submission questionnaire.

Once your full submission is complete, your paper will undergo a series of checks in preparation for peer review. After your manuscript has passed the checks it will be sent out for review. To provide the metadata for your submission, please Login to Editorial Manager (https://www.editorialmanager.com/pbiology) within two working days, i.e. by Sep 06 2023 11:59PM.

If your manuscript has been previously peer-reviewed at another journal, PLOS Biology is willing to work with those reviews in order to avoid re-starting the process. Submission of the previous reviews is entirely optional and our ability to use them effectively will depend on the willingness of the previous journal to confirm the content of the reports and share the reviewer identities. Please note that we reserve the right to invite additional reviewers if we consider that additional/independent reviewers are needed, although we aim to avoid this as far as possible. In our experience, working with previous reviews does save time.

If you would like us to consider previous reviewer reports, please edit your cover letter to let us know and include the name of the journal where the work was previously considered and the manuscript ID it was given. In addition, please upload a response to the reviews as a 'Prior Peer Review' file type, which should include the reports in full and a point-by-point reply detailing how you have or plan to address the reviewers' concerns.

During the process of completing your manuscript submission, you will be invited to opt-in to posting your pre-review manuscript as a bioRxiv preprint. Visit http://journals.plos.org/plosbiology/s/preprints for full details. If you consent to posting your current manuscript as a preprint, please upload a single Preprint PDF.

Feel free to email us at [email protected] if you have any queries relating to your submission.

Kind regards,

Christian

Christian Schnell, Ph.D.

Senior Editor

PLOS Biology

[email protected]

Decision Letter 1

Lucas Smith

3 Nov 2023

Dear Dr Peng,

Thank you for your patience while your manuscript "Phosphorylation of 53BP1 by ATM controls neurodevelopmental programs in cortical brain organoids" was peer-reviewed at PLOS Biology. It has now been evaluated by the PLOS Biology editors, an Academic Editor with relevant expertise, and by several independent reviewers. In light of the reviews, which you will find at the end of this email, we would like to invite you to revise the work to thoroughly address the reviewers' reports.

As you will see below, the reviewers appreciate the importance of the topic examined here and comment that the study offers some potentially interesting insights, but they have also highlighted that additional experimental work is needed to bolster the conclusions of the study. After discussion with the Academic Editor, we think these requests will need to be thoroughly addressed before we can consider your manuscript further for publication at PLOS Biology.

While we think it will be essential to expand the study, with new data and phenotypic analyses, we would not strictly require that you characterize an additional ES line as suggested by Reviewer 2. While we appreciate that adding these data would strengthen the study, we think that this request is beyond the scope of the current work. Additionally, we would not require the generation of new single cell RNA transcriptomics, but we do agree with the reviewers that more targeted analyses should be performed.

Given the extent of revision needed, we cannot make a decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is likely to be sent for further evaluation by all or a subset of the reviewers.

We expect to receive your revised manuscript within 3 months. Please email us ([email protected]) if you have any questions or concerns, or would like to request an extension.

At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not intend to submit a revision so that we may withdraw it.

**IMPORTANT - SUBMITTING YOUR REVISION**

Your revisions should address the specific points made by each reviewer. Please submit the following files along with your revised manuscript:

1. A 'Response to Reviewers' file - this should detail your responses to the editorial requests, present a point-by-point response to all of the reviewers' comments, and indicate the changes made to the manuscript.

*NOTE: In your point-by-point response to the reviewers, please provide the full context of each review. Do not selectively quote paragraphs or sentences to reply to. The entire set of reviewer comments should be present in full and each specific point should be responded to individually, point by point.

You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

2. In addition to a clean copy of the manuscript, please also upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Revised Article with Changes Highlighted" file type.

*Re-submission Checklist*

When you are ready to resubmit your revised manuscript, please refer to this re-submission checklist: https://plos.io/Biology_Checklist

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision:

*Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*PLOS Data Policy*

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5

*Blot and Gel Data Policy*

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Lucas

Lucas Smith, Ph.D.

Senior Editor

PLOS Biology

[email protected]

------------------------------------

REVIEWS:

Reviewer #1: This manuscript from Peng and colleagues investigate the role of ATM-dependent 53BP1 ser25 phosphorylation in neural development. Significantly, this work represents the follow up to prior work from the same group (Nature Neuroscience 2019), in which they reported a role for promoter bound 53BP1 - UTX complexes in upregulating neurodevelopmental genes during the differentiation of human embryonic stem cells into neurons or into cortical organoids.

Here the paper starts with the observation that 53BP1 expression doesn't change during differentiation from hESCs to neurons/cortical organoids, so they speculate it might instead be controlled by 53BP1 phosphorylation ATM, and then concentrate on the well-known Ser25-phosphosite in 53BP1 (a known ATM target) since this modification appears to be reduced in differentiated NPCs relative to hESCs in western blotted lysates. In correlation of such a notion, they find ATM-KO cells also show perturbed/modified transcriptional expression profiles during the differentiation experiments, they show ATM interacts with 53BP1 (by IP), and they show NPCs with 53BP1 Ser25Ala/Asp knockin mutations also have altered expression profiles, including alterations in neural gene expression profiles. Lastly, they correlate these changes to the altered residency of 53BP1 -S25 mutant proteins on specific gene promoters relative to wild type 53BP1 (which over-accumulates relative to WT), on which they speculate that ATM-dependent 53BP1 phosphorylation, guided by upstream DDR signalling incl ATM-dependent gH2AX induction at TSSs which recruits 53BP1, then negatively regulates 53BP1 occupancy on bivalent gene promoters, thereby modulating expression and contributing to neuronal differentiation.

The paper is quite descriptive and correlative in nature, with the bulk of data using transcriptomics and proteomics to identify changes in protein/gene expression between the different genetic backgrounds, GSEA/similar to attribute this to different gene classes that include (but are not limited to) genes involved in neural differentiation. In then showing that differences occur both in ATM and 53BP1 mutant cells, there may be some correlation/overlap, but it is difficult to interpret this as the stated causality. The authors identify interesting differences in S25A/D mutant 53BP1 vs WT 53BP1 promoter binding patterns, but how these alters gene expression patterns is hard to discern from the way the data is presented. I also found the evidence backing for the proposed model (and sequence of events) to be lacking. If ATM indeed regulates 53BP1 promoter binding via pS25 phosphorylation, this could be easily demonstrated by ChIP'ing wild type and mutant p53 in the ATM-KO cells, as one would expect the effect to increase promoter binding in the case of the wild-type but the mutant protein would not be impacted. However, even if this was the case, perhaps an even more important gap in the model is the DNA-damage stimulus for ATM-dependent regulation of 53BP promoter interactions? IT is proposed that DDR signalling via ATM be both positively and negatively regulates 53BP1-promoter interactions, via gH2AX-dependent recruitment, and Ser-25 phosphorylation of 53BP1, respectively. However, this is a counterintuitive concept to grasp, and since it is not supported by the included evidence, I'm left not really understanding the mechanism or how it might regulate neuronal identify or function.

Specific Comments:

- As described above, causality is not established in the case of how wild-type 53BP1 promoter interactions are impacted by ATM status, and to what degree this occurs via Ser-25 phosphorylation, thus this conclusion is not substantiated.

- Earlier work showed this 53BP1-depedentn regulation of neuronal function was not conserved between mouse and man. This manuscript should be transparent in stating this is not a well conserved mechanism.

- If this is all dependent on ATM signalling, what is then stimulating ATM activity, and how its this guiding this activity specifically at promoters?

- The author specuflate this might be controlled by gH2AX-dependent recruitment of 53BP1 (presumably to sites of DNA damage). If so, then this would be controlled by gH2AX-MDC1-RNF8-RNF168-H2AK15ub axis which is essential for 53BP1 chromatin binding. RNF168 KO cells would test or refute this hypothesis and such an experiment would be needed to substantiate such a claim

- Line 93-94: "By D55, a subset of ATM-KO cortical organoids appeared smaller than

controls, but this difference was not statistically significant (Supplementary Fig 3A-B). " - if it not significant this conclusion should be removed

- Line 33-36 "to localize to chromatin with double-stranded breaks, 53BP1 uses its BRCT domain to bind to gH2AX, the Tudor domain to bind to H4K27 dimethylation, and its UDR segment to bind to ubiquitinated H2AK15 (7, 8, 9, 36 10). " is incorrect; 53BP1 tudor recognise H4K20-methylation not this other state.

Reviewer #2: In the manuscript „Phosphorylation of 53BP1 by ATM controls neurodevelopmental programs in cortical brain organoids" by Lim et al., the authors generated ATM-KO brain organoids and found severe neural developmental defects in these organoids. Next, they generated hESC lines, in which 53BP1-S25 cannot be phosphorylated by ATM anymore, by mutating serine 25 to alanine or aspartic acid using CRISPR/Cas9. By generating brain organoids from these cell lines, the authors found that 53BP1-S25A and 53BP1-S25D brain organoids - while different in comparison to the ATM-KO organoids - exhibit abnormal neural development. Lastly, the authors identified direct target genes of 53BP1 by ChIP-seq. Thus, the authors provide mechanistic insight into the role of 53BP1 phosphorylation by ATM during neural development. As this study is very interesting and provides novel insight into the role of ATM and 53BP1 during neural development, it seems to be suitable for publication in PLOS Biology. However, I have some concerns, which need to be addressed before considering the manuscript for publication. Mainly, the phenotypic analysis of the ATM-KO, 53BP1-S25A and 53BP1-S25D brain organoids in terms of analyzed time points (1 to maximum 2 points depending on the mutation) and features/markers studied is not sufficient to draw the conclusions that the authors have drawn. A more thorough phenotypic analysis is needed to understand which neurodevelopmental processes are disturbed in these brain organoids and which contribution ATM and 53BP1 have in this process. For details, please see below.

1) Almost all analyses of brain organoids were performed at d35 with the exception of a few analyses at d55. On which basis has this time point of analyses been chosen? Brain organoids change during development in terms of morphology and cell type composition, so one or two time points of analyses are normally not sufficient. Could the authors please address the following questions? Was this the only time point when the phenotype was present or strongest? How do early-stage brain organoids look like? When did first abnormalities appear? For completeness, it would be good to add this information to the manuscript.

2) In line with the previous question, in d35 ATM-KO organoids, at least in the pictures shown in Figure 1G, no ventricle-like structures are visible. This raises the question, if these structures degenerated during brain organoid development or if they were never present?

3) The ATM-KO brain organoid exhibit many PAX6-positive cells but these cells are not (well) organized in VZ-like structures (not densely packed, no radial organization). This raises several questions. (i) How did the authors identify the VZ (size)? Would ZO-1 stainings helpful to identify the ventricular surface? (ii) Are the Pax6-positive cells apical radial glia (with an apical contact, here ZO-1 stainings could be helpful again) or did they differentiate to a different progenitor type (without apical contact; basal progenitors?)? (iii) It seems so that there are many more Pax6-positive cells in ATM-KO in comparison to WT brain organoids. Is this true? Could the authors please quantify Pax6-positive cells in WT and ATM-KO brain organoids? This seems to also be the case for Ki67. Could the authors also quantify Ki67? These data would be important to better understand the effects of ATM on neural development.

4) In line 92/93 authors claim to observe reduced neuronal differentiation. How do the authors know this? From the images presented in Figure 1G, it seems so that this is true for some ATM-KO clones but not for all. Could the authors validate this by quantifications. Moreover, the authors present just CTIP2 stainings, which is a marker for deep-layer neurons. Could it be that just deep-layer neurons are affected? Why not using a general neuronal marker like NeuN? Would upper-layer neurons at later stages also be affected?

5) How do the authors know that there are lower levels of proliferation in ATM-KO organoids? For this they would need to quantify the number of KI67 positive cells (see above). Information on mitotic (e.g. PH3 positive) cells would also be helpful, as apical radial glia normally divide at the ventricular surface. Is there reduced mitotic cells because there is no ventricular surface or can these cells now divide freely within in the organoid? ZO-1 stainings in combination with PH3 would be helpful.

6) WT and ATM-KO brain organoids seem to have a very different cell type composition. This might bias the transcriptome analyses: Instead of getting mechanistic insight in what ATM-KO is changing within a given cell population, one would get differences which are caused by different cell type composition. Here, single cell RNAseq or enrichment for certain cell populations (e.g. progenitors vs neurons) would be helpful. Another option would be to perform transcriptomics on NPCs, which the authors could easily produce from WT and ATM-KO cells.

7) Brain organoids and KO lines were derived from one cell line (H9/WA09). Would these phenotypes also be observed, if another cell line would be used in this study? Organoid studies normally using more than one cell line to address biological variation.

8) As ATM phosphorylates 53BP1 and therefore at least partially functions through 53BP1 phosphorylation, I would expect 53BP1-S25A and 53BP1-S25D to have similar phenotypes as ATM-KO. However, the phenotypes are quite different, e.g. number and organization of PAX6-positive cells. How do the authors interpret this? Could the authors please add their considerations to the discussion part?

9) The number of CTIP2-positive cells, from the pictures presented in Figure 3D, looks very different between the various 53BP1-S25A and 53BP1-S25D clones. Could the authors please quantify CTIP2-positive cell numbers and compare between 53BP1-S25A and 53BP1-S25D clones and WT?

10) Why are there fewer progenitor cells in 53BP1-S25A and 53BP1-S25D organoids? Do they proliferate less or have a longer cell cycle or is there increased apoptosis or increased neuronal differentiation? As already mentioned above the characterization of brain organoids is very limited and needs to be expanded to understand the effect of 53BP1-S25A and 53BP1-S25D on neural development, especially as the transcriptomic studies seem not provide a clear mechanism.

11) The transcriptome analysis of 53BP1-S25A and 53BP1-S25D brain organoids does not provide a clear explanation of the observed phenotypes. As described in comment 6) might this be due to the different cell type composition between 53BP1-S25A, 53BP1-S25D and WT organoids? Could the transcriptome analysis not also be performed on cultured NPCs, as this would be a clean population and as this would provide a direct comparison to the ChIP-seq data which was performed on these cells anyway?

Minor comments:

1) The legend of Figure 1A is missing.

2) How did the authors confirm that the generation of NPCs were successful?

3) While the authors have checked, if their CRISPR/Cas9-edited cells were still pluripotent, they have not checked, if the karyotype of these cells is still normal.

4) In Figure 1F and Supplemental Figure S2B, why is there a band visible for ATM in case of some ATM-KO cell lines?

5) In Figure 2E, what do the error bars indicate?

Reviewer #3: In this study, Lim, Djekidel and colleagues investigate the role of ATM and its target 53BP1 in cortical development. After confirming that 53BP1 is a target of ATM on residue S25, they generate a series of KO and KI ES cell lines for these factors and generate cerebral organoids. In both ATM KO and 53BP1 SA and SD, they observe major developmental defects, including proliferation, fate and overall growth. Because ATM expression and 53BP1 phosphorylation increase between ES cells and NPCs in the absence of increased gamma-H2AX, the authors propose that the observed function of 53BP1 phosphorylation is independent of DNA damage. They next use an extensive array of omics methods to better characterise these defects, including transcriptomics, proteomics, phospho-proteomics and ChIP-seq. These experiments identify a series genes and pathways altered by ATM and 53BP1 mutations and demonstrate that 53BP1 phosphorylation controls its position on chromatin.

While the study of ATM / 53BP1 during cortical development is certainly an important topic, the manuscript falls short of providing novel conceptual advance. The organoid defects are certainly clear but could arise from any type of defect, including organoid specification defects. The multi-omics analysis is impressive, but we are left with large numbers of altered genes and pathways, a lot of them probably as consequences of organoid defects, and none of this information is used to identify how ATM / 53BP1 may affect early neurogenesis.

Specific comments

1/ The authors suggest that the role of ATM / P-53BP1 during neurodevelopment could be independent of DNA damage, based on absence of increase of gamma-H2AX in NPCs, as compared to ES cells. However, for this suggestion to hold, DNA damage should be looked at in cerebral organoids where the phenotypic analysis was performed. Related to this, do the authors detect increased apoptosis in their organoid models (as suggested by their omics data).

2/ The fact that mutated organoids are altered is very clear and convincing, but it is hard to grasp what is causing the defect. The organoids are highly disorganized without clear luminal structures. Is organoid specification correct? In figure 3D for example, PAX6 staining appears severely reduced, which could reflect incorrect identity of the organoids.

3/ In figures 1 and 3, the quantification of the defects should be properly normalized. The fraction of PH3 or Ki67 cells should be quantified out of PAX6+ progenitors, in order to conclude whether progenitor proliferation is affected. For cell fate, it would be good to quantify the fraction of PAX6+ and Ctip2+ cells out of total cells (DAPI) or to present a PAX6/Ctip2 ratio.

4/ The authors should provide a ZO-1 staining (or similar) to quantify apical surface area and the formation (or absence of formation) of lumen.

5/ The PAX6 staining appears very different between ATM KO and 53BP1 KI conditions. Does this reflect different molecular alterations?

Decision Letter 2

Christian Schnell, PhD

4 Jul 2024

Dear Dr Peng,

Thank you for your patience while we considered your revised manuscript "Phosphorylation of 53BP1 by ATM controls neurodevelopmental programs in cortical brain organoids" for publication as a Research Article at PLOS Biology. This revised version of your manuscript has been evaluated by the PLOS Biology editors, the Academic Editor and the original reviewers.

Based on the reviews and on our Academic Editor's assessment of your revision, we are likely to accept this manuscript for publication, provided you satisfactorily address the remaining points raised by the reviewers. In particular, we share Reviewer 1's concerns about the strength of the data implicating HH. We think that it is beyond the scope of the manuscript to work out exactly how ATM is activated in this context, but that the interpretation be toned down, given that the conclusion is based on only a slight difference in the variance of the Western blot data that influences the statistics. Therefore, we suggest that you modify the text to reflect this uncertainty.

Please also make sure to address the following data and other policy-related requests.

* We would like to suggest a different title to improve accessibility: "Phosphorylation of the DNA damage repair factor 53BP1 by ATM kinase controls neurodevelopmental programs in cortical brain organoids"

* Please add the links to the funding agencies in the Financial Disclosure statement in the manuscript details

* DATA POLICY:

You may be aware of the PLOS Data Policy, which requires that all data be made available without restriction: http://journals.plos.org/plosbiology/s/data-availability. For more information, please also see this editorial: http://dx.doi.org/10.1371/journal.pbio.1001797

Note that we do not require all raw data. Rather, we ask that all individual quantitative observations that underlie the data summarized in the figures and results of your paper be made available in one of the following forms:

1) Supplementary files (e.g., excel). Please ensure that all data files are uploaded as 'Supporting Information' and are invariably referred to (in the manuscript, figure legends, and the Description field when uploading your files) using the following format verbatim: S1 Data, S2 Data, etc. Multiple panels of a single or even several figures can be included as multiple sheets in one excel file that is saved using exactly the following convention: S1_Data.xlsx (using an underscore).

2) Deposition in a publicly available repository. Please also provide the accession code or a reviewer link so that we may view your data before publication.

Regardless of the method selected, please ensure that you provide the individual numerical values that underlie the summary data displayed in the following figure panels as they are essential for readers to assess your analysis and to reproduce it:

1DHI, 2ABCE, 3E, 4CF, 5ABCEFGHI, S1I, S2E and similar panels in the supplementary figures

NOTE: the numerical data provided should include all replicates AND the way in which the plotted mean and errors were derived (it should not present only the mean/average values).

Please also ensure that figure legends in your manuscript include information on where the underlying data can be found, and ensure your supplemental data file/s has a legend.

Please ensure that your Data Statement in the submission system accurately describes where your data can be found.

* Per journal policy, if you have generated any custom code during the course of this investigation, please make it available without restrictions. Please ensure that the code is sufficiently well documented and reusable, and that your Data Statement in the Editorial Manager submission system accurately describes where your code can be found.

Please note that we cannot accept sole deposition of code in GitHub, as this could be changed after publication. However, you can archive this version of your publicly available GitHub code to Zenodo. Once you do this, it will generate a DOI number, which you will need to provide in the Data Accessibility Statement (you are welcome to also provide the GitHub access information). See the process for doing this here: https://docs.github.com/en/repositories/archiving-a-github-repository/referencing-and-citing-content

* BLOT AND GEL REPORTING REQUIREMENTS:

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare and upload them now. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

As you address these items, please take this last chance to review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the cover letter that accompanies your revised manuscript.

We expect to receive your revised manuscript within two weeks.

To submit your revision, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' to find your submission record. Your revised submission must include the following:

- a cover letter that should detail your responses to any editorial requests, if applicable, and whether changes have been made to the reference list

- a Response to Reviewers file that provides a detailed response to the reviewers' comments (if applicable, if not applicable please do not delete your existing 'Response to Reviewers' file.)

- a track-changes file indicating any changes that you have made to the manuscript.

NOTE: If Supporting Information files are included with your article, note that these are not copyedited and will be published as they are submitted. Please ensure that these files are legible and of high quality (at least 300 dpi) in an easily accessible file format. For this reason, please be aware that any references listed in an SI file will not be indexed. For more information, see our Supporting Information guidelines:

https://journals.plos.org/plosbiology/s/supporting-information

*Published Peer Review History*

Please note that you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*Press*

Should you, your institution's press office or the journal office choose to press release your paper, please ensure you have opted out of Early Article Posting on the submission form. We ask that you notify us as soon as possible if you or your institution is planning to press release the article.

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Please do not hesitate to contact me should you have any questions.

Sincerely,

Christian

Christian Schnell, PhD

Senior Editor

[email protected]

PLOS Biology

Reviewer remarks:

Reviewer #1: The manuscript remains descriptive, since the authors are still unable to explain how the ATM-gH2AX-RNF168-53BP1 is activated, and how/why this non-canonical role of DNA damage signaling is controlling specific genetic programs in NPCs and cortical organoids, while in other somatic tissues the gH2AX-RNF169-53BP1 pathway is a well-established to specifically regulate DNA repair. They have tried to address the activating stimulus of ATM, and in new experiments with cyclopamine (HH inhibitor) they find reduced pS25-53BP1 levels and suggest that in cortical neurons, hedgehog signalling is likely required to activate ATM. Again, HH would be an unexpected and non-canonical activator of ATM, and the claimed alterations of pS25-53BP1 levels in new immunoblot data in new Supp Fig 16 aren't clear-cut enough to convince me that these conclusions have sufficiently substantiated evidence basis. The author rebuttal states that "the exact clarification of mechanisms promoting ATM activities, especially in directing its kinase activity at specific promoters, is beyond the scope of this study. " yet nevertheless they also state that this study "has broad implications about diseases associated with ATM, including ataxia telangiectasia". Although, I acknowledge that the authors have attempted to address my concerns with new experiments to explore the role of gH2AX, and add new correlative data regarding RNF168, there absence of solid evidence to explain the mechanism basis of this non-canonical role of the ATM-gH2AX-53BP1 axis, and so I remain cautiously sceptical of the proposed mechanisms and their overall impact.

Reviewer #2: The authors addressed all my comments and improved the manuscript substantially. I can recommend the current version of the manuscript for publication in PLOS Biology.

Reviewer #3: The authors have now addressed all of my previous concerns: the absence of DNA damage increase with differentiation, the correct specification of mutant organoids (comments 1 and 5), the normalization method for the quantifications, and the use of ZO-1 staining to quantify the lumen defects.

Decision Letter 3

Christian Schnell, PhD

19 Jul 2024

Dear Dr Peng,

Thank you for the submission of your revised Research Article "Phosphorylation of the DNA damage repair factor 53BP1 by ATM kinase controls neurodevelopmental programs in cortical brain organoids" for publication in PLOS Biology. On behalf of my colleagues and the Academic Editor, Madeline Lancaster, I am pleased to say that we can in principle accept your manuscript for publication, provided you address any remaining formatting and reporting issues. These will be detailed in an email you should receive within 2-3 business days from our colleagues in the journal operations team; no action is required from you until then. Please note that we will not be able to formally accept your manuscript and schedule it for publication until you have completed any requested changes.

Please take a minute to log into Editorial Manager at http://www.editorialmanager.com/pbiology/, click the "Update My Information" link at the top of the page, and update your user information to ensure an efficient production process.

While you are attending to the formatting and production requests to come, we would also like you to state the limitations of the data shown in Fig S16 more clearly. We are not convinced that it is accurate to single out HH signaling, and it would be rather more accurate to simply say that the data suggest potential effects of those signaling pathways but more experiments would be needed.

PRESS

We frequently collaborate with press offices. If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximise its impact. If the press office is planning to promote your findings, we would be grateful if they could coordinate with [email protected]. If you have previously opted in to the early version process, we ask that you notify us immediately of any press plans so that we may opt out on your behalf.

We also ask that you take this opportunity to read our Embargo Policy regarding the discussion, promotion and media coverage of work that is yet to be published by PLOS. As your manuscript is not yet published, it is bound by the conditions of our Embargo Policy. Please be aware that this policy is in place both to ensure that any press coverage of your article is fully substantiated and to provide a direct link between such coverage and the published work. For full details of our Embargo Policy, please visit http://www.plos.org/about/media-inquiries/embargo-policy/.

Thank you again for choosing PLOS Biology for publication and supporting Open Access publishing. We look forward to publishing your study. 

Sincerely, 

Christian

Christian Schnell, PhD

Senior Editor

PLOS Biology

[email protected]

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Data. Numerical data used to generate summary data in this study.

    (XLSX)

    pbio.3002760.s001.xlsx (273.3KB, xlsx)
    S1 Raw Images. Uncropped western blot images in this study.

    (PDF)

    pbio.3002760.s002.pdf (988.2KB, pdf)
    S1 Fig. Characterization of 53BP1-pS25 and NPCs and genome editing of hESCs.

    (A) Schematic diagram of neural differentiation of hESCs: neural induction, differentiation, and maturation media to form EBs, rosettes, NPCs, and neurons. (B) Principal component analysis of WT ESCs, NPCs, day 10 (D10) cortical organoids, and D17 cortical organoids. GSEA terms that are highly enriched in significantly (C) down-regulated and (D) up-regulated genes in WT NPCs compared to ESCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. (E) Immunofluorescence of NPC markers PAX6 and NESTIN. Bar, 50 μm. (F) Quantification of 53BP1-pS25-positive hESCs or hNPCs. Data are presented as the mean ± SEM, with p < 0.0001. (G) WB analysis of control cells and 53BP1-KO clones 415, 416, and 209, which are clones KO1, KO2, and KO3 in Yang and colleagues’ study [6]. (H) WB analysis of control and 53BP1-S25A hNPCs. The S25A mutation prohibits phosphorylation. (I) WB analysis of hESCs and hNPCs and quantification. (J) Schematic diagram of genome editing in hESCs. Guide RNA 6 were complexed with Cas9 proteins and used along single-stranded nucleotide donors to transfect hESCs. Individual clones from transfection were cultured, sequenced by mi-seq across the targeted 53BP1 locus, and established as >99% pure clonal lines. Diagram was generated using open-sourced images available at biorender.com. Underlying numerical values for figures are found in S1 Data. EB, embyoid body; ESC, embryonic stem cell; GSEA, gene set enrichment analysis; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WB, western blot; WT, wild type; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

    (PDF)

    pbio.3002760.s003.pdf (445.8KB, pdf)
    S2 Fig. Generation and analyses of ATM-KO hESCs and cortical organoids.

    (A) Alignment of WT and ATM-KO mutation sequences on 2 alleles (al) in the ATM locus. Red indicates the gRNA sequence. (B) WB analysis of WT and 4 ATM-KO hNPCs. (C) Principal component analysis showed the intermixing and similar RNA-seq profiles from hESCs of 7 WT, 4 53BP1-S25A, 4 53BP1-S25D, 4 ATM-KO, and 4 53BP1-KO lines. (D) Immunofluorescence showed similar expression of OCT4 and SSEA4 proteins in control and ATM-KO hESCs. Bar, 100 μm. (E) WB analysis of WT and 2 ATM-KO hNPCs. Quantification suggests reduction of γH2AX in ATM-KO hNPCs. Welch’s t test was used to perform pairwise comparisons of WT and ATM-KO. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; KO, knockout; WB, western blot; WT, wild type.

    (PDF)

    pbio.3002760.s004.pdf (385.8KB, pdf)
    S3 Fig. Analysis of γH2AX and CC3 in cortical organoids.

    (A) Immunofluorescence showed D35 ATM-KO and WT cortical organoids had similar γH2AX foci. Bar, 100 μm. FACS analysis of CC3 in (B) D21 and (C) D28 cortical organoids. Two biological replicates were done, and each data point was based on 3 technical replicate analyses of 10–12 cortical organoids. (D, E) Immunofluorescence and quantification of CC3 in D28 cortical organoids. Bar, 100 μm. Graphs are presented in ratios (out of 1), with **, p < 0.01; ****, p < 0.0001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; CC3, cleaved-caspase 3; KO, knockout; WT, wild type.

    (PDF)

    pbio.3002760.s005.pdf (207.8KB, pdf)
    S4 Fig. Analysis of cell proliferation in cortical organoids.

    FACS analysis of PAX6 and KI67 in (A, B) D28 and (C) D35 cortical organoids. Each data point was based on the 3 technical replicate analyses of 10 cortical organoids. (D) Quantification of KI67/PAX6 ratios in immunofluorescence of D35 cortical organoids. Each data point represents quantification of cells in 1 cortical organoid. (E, F) Immunofluorescence and quantification of H3-pS10 (PH3) in D28 cortical organoids. Bar, 100 μm. (F-H) Immunofluorescence and quantification of PH3 and KI67 in D35 cortical organoids. Bar, 100 μm. ***, p < 0.001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data.

    (PDF)

    pbio.3002760.s006.pdf (338.2KB, pdf)
    S5 Fig. Immunofluorescence analyses of cortical organoids and NPCs.

    Immunofluorescence of (A) NEUN and (E) ZO-1 and PAX6 in D37 cortical organoids. Bar, 100 μm. (B) Immunofluorescence of ZO-1 in D28 cortical organoids. Bar, 100 μm. Quantification of the (C) number and (D) surface area of ZO-1-positive ventricles in D28 cortical organoids. *, p < 0.05; ***, p <0 .001; ns, not significant by two-way ANOVA test. (F) Bright-field images of cortical organoids formed by ATM-KO2, 3, 14, 43, and WT control at day 55 of differentiation. Bar, 1.5 mm. (G) The size of cortical organoids was compared between groups by one-way ANOVA with Dunnett’s multiple comparisons test, with ns, not significant and ***, p < 0.001. n = 13 organoids/group. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; NPC, neural progenitor cell; WT, wild type.

    (PDF)

    pbio.3002760.s007.pdf (389.3KB, pdf)
    S6 Fig. Characterization of NPCs and D35 cortical organoids.

    (A) Immunofluorescence of PAX6 and NES in NPCs. Bar, 50 μm. (B) Principal component analysis of proteomics data of D35 WT and ATM-KO cortical organoids. GSEA terms that are highly enriched in significantly (C) higher and (D) lower total proteins in D35 ATM-KO versus WT cortical organoids. (E) GSEA terms that are highly enriched in significantly higher phosphoproteins, which were normalized to total proteomics, in D35 ATM-KO versus WT cortical organoids. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type.

    (PDF)

    pbio.3002760.s008.pdf (309.8KB, pdf)
    S7 Fig. Kinase activities in cortical organoids and characterization of the 53BP1-S25A and 53BP1-S25D hESCs.

    Heatmaps showing relative phosphorylation levels of (A) 7 MAPK9 substrates that are significantly lower and (B) 7 CDK5 substrates that are significantly higher in D35 ATM-KO versus WT cortical organoids. (C) Heatmaps showing activity of selected protein kinases between ATM-KO3, ATM-KO4, and WT cell lines. (D) Alignment of WT and 53BP1-S25A and S25D mutation sequences on 2 alleles (al). Red indicates the gRNA sequence. Underline indicates codon encoding the WT serine 25, mutant alanine, or mutant aspartic acid. (E) WB analysis of control and 53BP1-S25D hNPCs, which have comparable levels of 53BP1 protein. (F) Transcripts per million values of 10 pluripotency genes were used for comparison to show that control, 53BP1-S25A, and 53BP1-S25D hESCs did not differ in pluripotency. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; hESC, human embryonic stem cell; hNPC, human neural progenitor cell; KO, knockout; WB, western blot; WT, wild type; 53BP1, p53 binding protein 1.

    (PDF)

    pbio.3002760.s009.pdf (393.5KB, pdf)
    S8 Fig. Characterization of the 53BP1-S25A and 53BP1-S25D hESCs and cortical organoids.

    (A) Immunofluorescence showed similar expression of OCT4 and SSEA4 proteins in WT, 53BP1-S25A, and 53BP1-S25D hESCs. Bar, 100 μm. Immunofluorescence of (B) KI67 and (D) PH3 in cryosections of cortical organoids at day 35 of differentiation. Bar, 100 μm. (C) Quantification of KI67-positive cells in D35 cortical organoids. Data points represent single organoids. The mean ± SEM values were compared by one-way ANOVA with Dunnett’s multiple comparisons test to yield ****, ***, and ** indicating p < 0.0001, 0.001, and 0.01, respectively. n = 3 organoids/group. Underlying numerical values for figures are found in S1 Data.

    (PDF)

    pbio.3002760.s010.pdf (402.8KB, pdf)
    S9 Fig. Analysis of γH2AX and CC3 in cortical organoids.

    Immunofluorescence of (A) γH2AX in D35 cortical organoids and (D) CC3 in D28 cortical organoids. Bar, 100 μm. CC3 quantification by FACS of (B) D21 and (C) D28 cortical organoids. For each datapoint, 10–12 organoids from each line were analyzed via 3 technical replicates, and data from 4 mutant lines were consolidated to achieve rigorous comparisons. **, p < 0.01 and ns, not significant by two-way ANOVA test. (E) CC3 quantification of immunofluorescence images of D28 cortical organoids. For each line, 4–6 images and >10,000 cells were analyzed. *, p < 0.05; **, p < 0.01; ns, not significant by two-way ANOVA test. Graphs in (B, C, E) are presented in ratios (out of 1). Underlying numerical values for figures are found in S1 Data.

    (PDF)

    pbio.3002760.s011.pdf (333.1KB, pdf)
    S10 Fig. 53BP1-pS25 promotes ventricle formation in cortical organoids.

    Quantification of the (A) number and (B) surface area of ZO-1-positive ventricles. (C) Immunofluorescence of ZO-1 in D28 cortical organoids. Bar, 100 μm. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ns, not significant by two-way ANOVA test. Underlying numerical values for figures are found in S1 Data.

    (PDF)

    pbio.3002760.s012.pdf (268.4KB, pdf)
    S11 Fig. Characterization of 53BP1-S25A and 53BP1-S25D cortical organoids.

    (A) Bright-field images of cortical organoids formed by cell lines 53BP1-S25A 34–3, 34–4, 79–1, 79–3 and S25D 14–3, 14–15, 14–19, 17, and 2 WT control at day 55 of differentiation. Bar, 1.5 mm. Blue transparent structures around organoids are Matrigel embedment. (B) At day 55 of differentiation, the size of cortical organoids was compared between groups. (C) The growth (comparing organoids at days 35 and 55) of cortical organoids were compared between groups. Data points represent single organoids. The mean ± SEM values were compared by one-way ANOVA with Dunnett’s multiple comparisons test to yield **** and ** indicating p < 0.0001 and 0.01, respectively. n = 15–36 organoids/group. (D) Two genes overlapped between up-regulated genes in 53BP1-S25A versus WT and down-regulated genes in 53BP1-S25D versus WT cortical organoids. No gene overlapped between down-regulated genes in 53BP1-S25A versus WT and up-regulated genes in 53BP1-S25D versus WT cortical organoids. (E) Down-regulated GSEA terms between 53BP1-S25A versus WT and 53BP1-S25D versus WT were not highly overlapped. Ten GSEA terms were specific to 53BP1-S25D versus WT. Underlying numerical values for figures are found in S1 Data.

    (PDF)

    pbio.3002760.s013.pdf (457.1KB, pdf)
    S12 Fig. Characterization of NPCs and comparative analyses of RNA-seq data.

    (A) Immunofluorescence of NPC markers PAX6 and NESTIN. Bar, 50 μm. GSEA identified top enrichment of differentially expressed genes in (B, D) 53BP1-S25A or (C, E) S25D versus WT NPCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. Venn diagrams depict overlaps between down-regulated genes in ATM-KO with 53BP1- (F) S25A or (G) S25D cortical organoids. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type.

    (PDF)

    pbio.3002760.s014.pdf (413.2KB, pdf)
    S13 Fig. Comparisons of RNA-seq data and 53BP1 ChIP-seq analyses.

    (A) GSEA graphs showed that up-regulated genes in 53BP1-S25A or S25D vs. WT had significant enrichment in down-regulated genes of ATM-KO vs. WT cortical organoids. P values were calculated by the hypergeometric test, assuming normal data distribution. (B) Concordantly differential expression of genes in 53BP1-S25D vs. WT were enriched in those in 53BP1-S25A vs. WT. (C) Concordantly differential expression of genes in 53BP1-S25A vs. WT were enriched in those in 53BP1-S25D vs. WT. For (A-C), P values were calculated by the hypergeometric test, assuming normal data distribution. (D) Proportions of 53BP1 binding to genomic features. 53BP1 ChIP-seq tracks at loci of representative (E) up-regulated and (F) down-regulated genes in 53BP1-S25A and S25D versus WT D35 cortical organoids. (G) S25A and S25D down-regulate 53BP1 targets that are enriched in IRE1-mediated unfolded protein response, regulation of cellular response to stress, iron import into cells, and regulation of apoptosis. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; WT, wild type.

    (PDF)

    pbio.3002760.s015.pdf (530.4KB, pdf)
    S14 Fig. 53BP1 ChIP-seq and 53BP1-pS25 CUT&RUN.

    (A) MA plot displays 53BP1 ChIP-seq signals at genomic sites that are significantly different in ATM-KO vs. WT NPCs. Proportions of genomic features and gene ontology of genes with (B) higher or (C) lower 53BP1 binding in ATM-KO vs. WT NPCs. (D) Proportions of 53BP1-pS25 binding to genomic features. (E) GSEA identified top enrichment of genes occupied by 53BP1-pS25 in WT NPCs. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; GSEA, gene set enrichment analysis; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; WT, wild type.

    (PDF)

    pbio.3002760.s016.pdf (222KB, pdf)
    S15 Fig. Differential 53BP1 ChIP-seq in 53BP1-WT, S25A, and S25D NPCs.

    (A) Principal component analysis of top 3,000 most variable peaks in 53BP1 ChIP-seq of 53BP1-WT, S25A, and S25D NPCs. Two independent cell lines for each group were used for ChIP-seq. Proportions of genomic features in regions with significantly different 53BP1 ChIP-seq in (B) 53BP1-S25A vs. WT and (C) 53BP1-S25D vs. WT, using the criterion of FC>2 and p < 0.05. (D) Heatmaps aligning peaks with significantly different 53BP1 ChIP-seq in 53BP1-S25A vs. S25D. Control regions are those, after voom normalization, showed the least changes and served as semi-independent validation of differential ChIP-seq analysis. Bubble graphs present top enriched categories of genes that had significantly lower 53BP1 ChIP-seq in (E) 53BP1-S25A vs. WT and (F) 53BP1-S25D vs. WT. Underlying numerical values for figures are found in S1 Data. FC, fold-change; NPC, neural progenitor cell; WT, wild type.

    (PDF)

    pbio.3002760.s017.pdf (297.8KB, pdf)
    S16 Fig. Analysis of ATM activities during the inhibition of TGFβ, WNT, and HH signaling.

    (A) Schematic diagram of neural specification of hESCs with HH (SB421542), TGFβ (dorsomorphin), and WNT (IWR1e and cyclopamine) signaling inhibitors. Nuclear extract was harvested on day 4 and day 10. (B) WB analysis of day 4 samples. (C) Quantification of day 4 WB. Data are presented as the mean ± SEM, and Student t test was performed for pairwise comparisons. n.s., *, and ** indicate not significant, p < 0.05, and p < 0.01, respectively. (D) WB analysis of day 10 samples. (E) Quantification of day 10 WB. Data are presented as the mean ± SEM, and Student t test was performed for pairwise comparisons. n.s., *, and ** indicate not significant, p < 0.05, and p < 0.01, respectively. Underlying numerical values for figures are found in S1 Data. ATM, ataxia telangiectasia mutated; hESC, human embryonic stem cell; WB, western blot.

    (PDF)

    pbio.3002760.s018.pdf (281.7KB, pdf)
    S17 Fig. RNF168-KO alters key genetic programs and 53BP1-pS25 binding to chromatin.

    (A) Alignment of WT and RNF168-KO mutation sequences in the RNF168 locus. Red indicates the gRNA sequences. (B) WB analysis of WT and RNF168-KO hESCs. (C) RT-qPCR analysis showing that pluripotent genes in RNF168-KO were expressed higher or the same as those in WT. RNF168-KO did not reduce pluripotent gene expression. *, p < 0.05; ns, not significant by two-way ANOVA text. (D) Immunofluorescence showed similar expression of OCT4 and SSEA4 proteins in WT and RNF168-KO hESCs. Bar, 100 μm. (E) Immunofluorescence of showed similar expression of PAX6 and NES in NPCs. WT and RNF168-KO NPCs. Bar, 50 μm. Functional terms that are highly enriched in (F) up-regulated and (G) down-regulated genes in RNF168-KO D35 cortical organoids. % Match, % of genes in the enriched term that overlap the differentially expressed genes or proteins. (H) Heatmaps aligning peaks with 53BP1-pS25 CUT&RUN signals that were gained, the same, or lost in RNF168-KO vs. WT NPCs, using the criterion of FC>2 and p < 0.05. n = numbers of peaks. Regions with the same signals, are n = 899, which showed the least changes after voom normalization and served as semi-independent validation of differential ChIP-seq analysis. (I) Functional terms of 53BP1-pS25-bound genes in WT NPCs. % Match, % of genes in the enriched term that overlap the differentially bound genes. (J) Number of differentially expressed genes identified by comparison of RNF168-KO vs. WT NPCs at p < 0.05. Of these genes, we list the numbers of 53BP1-pS25-bound targets and targets with higher or lower 53BP1-pS25 CUT&RUN signals in RNF168-KO NPCs. Underlying numerical values for figures are found in S1 Data. FC, fold-change; hESC, human embryonic stem cell; KO, knockout; NES, normalized enrichment score; NPC, neural progenitor cell; RT-qPCR, quantitative reverse transcription PCR; WB, western blot; WT, wild type; 53BP1-pS25, 53BP1 phosphorylated at serine 25.

    (PDF)

    pbio.3002760.s019.pdf (568.5KB, pdf)
    S1 Table. All cell lines generated for this study were treated with trypsin and Wright’s stain and then analyzed by the Cytogenetic Shared Resource at St. Jude.

    Typically normal karyotypes and 3 abnormalities are shown.

    (PDF)

    pbio.3002760.s020.pdf (1.1MB, pdf)
    S2 Table. The expression of forebrain, midbrain, and hindbrain markers in D35 WT and ATM-KO cortical organoids.

    Data suggest that D35 ATM-KO cortical organoids specified to the forebrain lineage.

    (PDF)

    pbio.3002760.s021.pdf (37.2KB, pdf)
    S3 Table. List of phosphoproteins, normalized to total protein levels, that were significantly lower in D35 ATM-KO versus WT cortical organoids.

    (XLSX)

    pbio.3002760.s022.xlsx (22.9KB, xlsx)
    S4 Table. Normalized (to total proteome) levels of phosphopeptide substrates of MAPK9 and CDK5 in D35 WT and ATM-KO cortical organoids.

    (XLSX)

    pbio.3002760.s023.xlsx (172.7KB, xlsx)
    S5 Table. Two-sample t test examines the sizes of cortical organoids that change between day 35 and day 55 of differentiation.

    Data from WT and 53BP1 mutants are compared pairwise by using the two-sample t test. The sizes of organoids are significantly different between each comparison pair (all p < 0.05).

    (PDF)

    pbio.3002760.s024.pdf (163.4KB, pdf)
    S6 Table. The changes in organoid size at days 35 and 55 of differentiation were compared to yield S3C Fig.

    This table lists the calculation for different combinations of data and the descriptive statistics.

    (PDF)

    pbio.3002760.s025.pdf (150.4KB, pdf)
    S7 Table. Primary antibodies used in this study.

    (PDF)

    pbio.3002760.s026.pdf (21.1KB, pdf)
    S8 Table. gRNA sequences used in this study.

    (PDF)

    pbio.3002760.s027.pdf (14.3KB, pdf)
    Attachment

    Submitted filename: response.pdf

    pbio.3002760.s028.pdf (16.4MB, pdf)
    Attachment

    Submitted filename: response.pdf

    pbio.3002760.s029.pdf (23.9KB, pdf)

    Data Availability Statement

    All sequencing data are deposited in NCBI GEO database under accession number GSE231321. Codes for analyzing sequencing data are deposited in https://doi.org/10.6084/m9.figshare.7411835. Mass spectrometry data were deposited in ProteomXchange, with project accession number PXD041699. Numerical data are in S1 Data, and uncropped Western blot images are in S1 Raw Images.


    Articles from PLOS Biology are provided here courtesy of PLOS

    RESOURCES