Summary:
Chromatin is a crucial regulator of gene expression and tightly controls development across species. Mutations in only one copy of multiple histone genes were identified in children with developmental disorders characterized by microcephaly, but their mechanistic roles in development remain unclear. Here we focus on dominant mutations affecting histone H4 lysine 91. These H4K91 mutants form aberrant nuclear puncta at specific heterochromatin regions. Mechanistically, H4K91 mutants demonstrate enhanced binding to the histone variant H3.3, and ablation of H3.3 or the H3.3-specific chaperone DAXX diminishes the mutant localization to chromatin. Our functional studies demonstrate that H4K91 mutant expression increases chromatin accessibility, alters developmental gene expression through accelerating pro-neural differentiation, and causes reduced mouse brain size in vivo, reminiscent of the microcephaly phenotypes of patients. Together, our studies unveil a distinct molecular pathogenic mechanism from other known histone mutants, where H4K91 mutants misregulate cell fate during development through abnormal genomic localization.
Keywords: developmental disorders, microcephaly, histone H3 variant H3.3, abnormal genomic localization, aberrant nuclear puncta, heterochromatin, neural differentiation
Introduction:
Chromatin integrates intrinsic cellular and environmental cues to regulate diverse DNA-templated processes, including replication, transcription, and DNA damage repair. Mutations in chromatin regulators, including nucleosome remodelers, writers, readers, and erasers of histone and DNA modifications, have been emerging in cancer and developmental disorders1,2. Histones are the main proteins in chromatin, which together with DNA form nucleosomes and higher-ordered chromatin structures. Four core histone proteins, H2A, H2B, H3, and H4, are encoded by multiple paralogous genes each to meet the demand for histones arising during DNA replication in S-phase. Somatic missense mutations located in all four core histones were recently discovered in malignancies, which changed the chromatin signatures of cancer and highlighted the importance of histones in human disease3–9. Mechanistic studies of recurrent histone H3 mutations in pediatric cancers revealed that mutated histones can inhibit the enzymatic activity of the cognate histone methyltransferases, and lead to changes in the epigenetic landscape and gene expression10–13.
In addition to cancer-associated histone mutations, de novo germline mutations in genes encoding histone H4 and histone H3 variant H3.3 have recently been identified in 33 and 59 patients, respectively, with developmental disorders, suggesting histones are key regulators in diseases beyond cancer14–20. Patients bearing these de novo heterozygous mutations share a phenotype of developmental delay and intellectual disability. While several somatic histone mutations have been well studied in oncogenesis, the mechanistic and functional roles of these germline histone mutations in human development remain largely unknown. A recent study on germline mutations changing H3.3 glycine 34 to arginine (G34R) identified that H3.3 G34R decreases the methylation of the adjacent K36 residue and impairs recruitment of a DNA methyltransferase to alter the expression of immune and neuronal genes, which contribute to progressive microcephaly and neurodegeneration20. Almost all studies on disease-associated histone mutants to date have focused on histone H3.3, yet whether and how other germline histone mutations dysregulate gene expression and impact development is unclear.
Here our study focuses on mutations affecting lysine 91 of histone H4, which to date have been reported in seven patients presenting with microcephaly, intellectual disability, and dysmorphic facial features14–16. In all these cases, patients carry mutations in just one out of the fourteen H4 gene copies in the human genome, and these mutations change lysine 91 to either glutamine (Q), arginine (R), or glutamic acid (E) (Figure S1A). H4K91 is located in the globular domain of H4 and at the H2B-H4 dimer-tetramer interface of nucleosomes (Figure S1B). Consistent with this important structural localization within the nucleosome, mutated H4K91 destabilizes nucleosomes in vitro, as inferred by more active H2A/H2B dimer exchange21. Yet how mutations of the H4 histone fold impact development is not clear. In this study, we aim to characterize the binding properties of H4K91 mutants, their genomic localization, and their roles in regulating gene expression as well as brain development to reveal key underlying mechanisms of patient-associated H4 germline mutations. Our in-depth study on the dominant H4 mutants unveils that histone mutants act through abnormal genomic localization, which to our knowledge defines a novel molecular pathogenic mechanism compared to previous research that focused on H3.3 mutations affecting histone posttranslational modifications (PTMs). Furthermore, our study provides a compelling molecular connection between H4 mutants and the histone variant H3.3, contributing to the intricate regulatory landscape of the histone mutations in developmental disorders.
Results:
H4K91 mutants display puncta formation and enhanced association with H3.3
The H4 protein is encoded by fourteen paralog genes, and heterozygous mutations of just one gene cause developmental defects in patients, suggesting that H4K91 mutants may have dominant effects on development. Therefore, we reasoned that expression of mutated H4 in the presence of endogenous wild-type H4 would recapitulate its functional effects, similar to effects observed with other histone mutations10–13,22. To characterize the function of mutated H4 during development, we first stably expressed HA-tagged wild-type H4 (H4WT) or H4K91 mutants in mESCs (Figure S1C). We focused subsequent studies on H4K91R and H4K91Q mutants as both are more prevalent in patients and more robustly accumulated in our model. Surprisingly, we found that mutated H4 appear as distinct nuclear puncta. In contrast, H4WT is homogeneously distributed within the nucleus (Figure 1A, Figure S1D). Quantification of the puncta intensity in cells with a wide range of expression levels confirmed that both H4 mutants, but not the wild-type H4, form distinct puncta (Figure 1B). Additionally, we observed the formation of discrete puncta in both live (Video S1) and fixed mESCs that transiently express GFP-tagged H4 mutants (Figure S1E), ruling out the possibility that the puncta identified by HA staining are caused by fixation and immunofluorescence staining23.
To understand the puncta formation of mutated H4, we sought to characterize its chromatin deposition pathway. During nucleosome assembly, H4 is deposited onto DNA within the H3/H4 tetramer, followed by the association of two H2A/H2B dimers. H4 is the only invariant core histone and thus binds to both canonical H3, deposited by CAF1, and the specialized histone variant H3.3, deposited in euchromatin regions by Hira, and in repetitive heterochromatic regions by ATRX and DAXX chaperones24–28. Because of the fundamental importance of the H3/H4 interaction in forming the nucleosome, we first asked whether H4K91 mutants can bind to both H3 and H3.3. We tested this by mononucleosome immunoprecipitation using HEK293T cells expressing HA-tagged H4WT or H4K91 mutants, followed by mass spectrometry. We found that H4K91 mutants preferentially associate with H3.3 over canonical H3, displaying a robust five-fold increase compared to WT H4 (Figure 1C–D). These results were further validated by western blotting with an H3.3-specific antibody (Figure 1E). Previous studies characterized H3.3 puncta in mESCs arising from deposition into heterochromatic regions, including telomeres 29–31. Consistent with increased binding to H3.3, H4K91 mutant puncta colocalized with the telomere probe and mutated H4-containing nucleosomes carried higher K9-trimethylated histone H3 (H3K9me3), a canonical heterochromatin mark (Figure S1E, F).
As the heterochromatin deposition of H3.3 is facilitated by the chaperone protein ATRX and DAXX, we investigated the localization of ATRX and DAXX in mESCs expressing mutated H425,31. Notably, these H4K91 mutant nuclear puncta co-localize with DAXX and ATRX (Figure 1F). To probe whether histone variant H3.3 and these chaperones are essential for H4 mutant puncta formation, we, expressed mutated H4 in mESC lines lacking H3.3, DAXX, or Hira, and analyzed H4 mutant localization. Staining and quantification of the puncta revealed that depletion of H3.3 or DAXX, but not the euchromatin-specific H3.3 chaperone HIRA, abolished the H4K91 mutant puncta formation in mESCs (Figure 1G, H). Altogether, these data show that H4K91 mutants are preferentially associated with H3.3 and deposited into heterochromatin by a DAXX/ATRX-dependent pathway.
H4K91 mutants are deposited into H3.3 and H3K9me3-enriched heterochromatin
DAXX and ATRX cooperate to deposit the H3.3/H4 complex into several distinct heterochromatic regions, including telomeres, pericentromeric regions, and interspersed repeats31–34. To define the incorporation sites of mutated H4 in mESCs, we profiled the genome-wide distribution of mutated H4K91 using chromatin immunoprecipitation followed by high-throughput DNA sequencing (ChIP-seq). After clustering H4K91 peaks based on the H4 ChIP signal, we found that both H4K91 mutants occupied a shared subset of new genomic loci (referred to as “K91 mutant-enriched peaks”), compared to H4WT. Interestingly, while H4K91Q localization was primarily confined to these shared mutant-enriched peaks, H4K91R was also localized to additional areas that also contain H4WT (referred to as “other peaks”), suggesting a wider deposition of the arginine mutant (Figure S2A). Further annotation of the shared K91 mutant-enriched peaks revealed that the majority (55%) of these peaks localized at distal intergenic regions, whereas most (52%) other peaks are at promoter regions (Figure S2B). We reasoned that the disease-causing function of H4 mutants is mediated by ectopic localization to de novo occupied regions shared across all mutants, represented by K91 mutant-enriched peaks. A closer examination of these peaks found that they have abundant H3.3 signal as well as H3K9me3, a marker of heterochromatin (Figure 2A, Table S1). Using the regions under both H4WT and H4K91 mutant peaks, we found that H4K91 mutant ChIP signal had a strikingly higher correlation with H3.3 and H3K9me3 ChIP signals than H4WT (Figure 2B), further corroborating the enhanced binding of mutant H4 to H3.3 documented in the pulldown assay (Figure 1D,E). Notably, K91 mutant-enriched peaks showed enrichment at several selected targets of H3.3 and H3K9me3, which were identified previously in mESCs (Figure 2C)32,33.
Next, we asked if the loss of H3.3 or its specific chaperone DAXX would blunt ectopic H4 enrichment, similar to the rescue of puncta in the knockout (KO) mESCs (Figure 1G, H). We found that K91 mutant-enriched peaks were diminished in H3.3 or DAXX KO mESCs but are maintained in Hira KO mESCs (Figure 2D). This is confirmed by the loss of genome-wide correlation of H4K91R or H4K91Q occupancy with H3.3-enriched sites (Figure S2C). Collectively, these data suggest that H4K91 mutants are incorporated into distinct genomic loci enriched in H3.3 and H3K9me3, and this ectopic recruitment relies on the histone variant H3.3 and its chaperone DAXX.
The observation of distinct puncta in the nucleus and distinct H3.3-dependent genomic localization of mutated H4 prompted us to study the potential effects of mutant expression on chromatin structure. Lysine 91 of H4 localizes at the H2B/H4 dimer tetramer interface, and this residue forms a salt bridge with an E76 residue in histone H2B (Figure S1B). H4K91 mutants are known to destabilize the nucleosome in vitro and in yeast21,35. Moreover, histone mutants affecting another H2B/H4 dimer-tetramer interface residue, H2B E76K, have been reported to destabilize nucleosomes in vitro and significantly promote chromatin accessibility8,9,21. To test whether disease-associated H4K91R/Q mutants alter chromatin accessibility in mammalian cells, we visualized accessible DNA using a previously reported assay of transposase-accessible chromatin with visualization (ATAC-see)36. Using the HDAC inhibitor Panobinostat as a positive control, we observed foci of increased accessibility in HEK293T cells that express K91 mutants, but not wild type control (Figure S3A–C). Altogether, we found that expression of patient-associated H4K91 mutants increases chromatin accessibility in mammalian cells.
H4K91 mutants alter mammalian brain development
We next investigated how the incorporation of H4K91 mutants could misregulate development. Patients carrying H4K91 mutations present with microcephaly, likewise an ectopic expression of H4K91 mutants (K91R/Q) in zebrafish led to underdeveloped brain15, together suggesting that a fraction of mutated H4 is sufficient to affect brain development in vivo. We sought to investigate the effect of H4K91 mutants on brain development in a mammalian system while minimizing the effects of genetic background, age, and sex. To achieve this, we generated H4WT and H4K91 mutant isogenic all-ESC mice using a tetraploid complementation approach. Specifically, we injected mESCs expressing HA-tagged H4 into tetraploid blastocysts, where the 4n cells of the host embryo contribute solely to the placenta, while the injected ESCs form the embryo proper37. H4K91 mutant pups at postnatal day 0 (P0) exhibited a reduction in brain size compared to H4WT mice, with no significant changes in body weight (Figure 3A). This phenotype mimics the microcephaly phenotypes observed in patients, suggesting that mice are a suitable model to study the underlying molecular mechanisms of H4K91 mutations. To visualize the localization pattern of WT and mutated H4 in vivo, we stained cerebral cortex sections with anti-HA antibody. Remarkably, H4 mutants show a similar pattern of nuclear puncta, and the puncta colocalize with DAPI-bright heterochromatin regions (Figure 3B). These findings recapitulate the distinct puncta detected in mESCs (Figure 1A, F, Figure S1D–E), suggesting that H4K91 mutants are consistently incorporated into heterochromatin in differentiated neurons in vivo.
In the cerebral cortex, neuronal progenitor cells localize to the ventricular and subventricular zone (SVZ), and undergo proliferation, differentiation, and migration to give rise to neurons and glial cells, providing a system to study lineage specification and cell differentiation38. To investigate the effect of H4K91 mutant expression in neurogenesis, we performed in situ hybridization with two probes targeting intermediate progenitor cells (Tbr2) and post-mitotic neurons (Satb2). In both wild type control and mutant cerebral cortex, we observed Tbr2 labeled progenitor cells localized at SVZ, while Satb2 labels cells in the upper cortical layers (Figure 3C). We quantified the relative thickness of Tbr2 or Satb2 positive cells normalized to full cortex thickness in different section areas from independent mice. These analyses revealed that the H4 mutant brain has a thinner Tbr2+ layer and expanded Satb2+ layer compared to H4WT (Figure 3D). These changes indicate that mutated H4 misregulates neurogenesis in vivo, resulting in a reduced progenitor population.
Accelerated neural differentiation occurs upon expression of H4K91 mutants
The H4K91 mutant-dependent phenotypes observed in mice mirror those of H3.3 loss, which promotes premature neural differentiation and altered gene expression in postmitotic neurons39,40. Since H4K91 mutants are recruited to H3.3-enriched genomic regions and are preferentially associated with H3.3 (Figure 1D, E, Figure 2A–B), we reasoned that H4 mutants, similar to the loss of H3.3, might misregulate neuronal differentiation and alter gene expression in differentiated neural cells. Therefore, we next investigated the effects of H4 mutants on neural differentiation. We differentiated mESCs to neural cells and detected Sox2+ NPCs and Map2+ neuron cells by immunostaining, indicating successful differentiation into the neuronal lineage (Figure S4A–B). We next asked whether H4 mutant expression alters gene expression by comparing transcriptional programs of four-day differentiated neural cells that express H4WT or mutant H4. We found overlapping transcriptional changes caused by H4K91R and H4K91Q mutants, further demonstrating that H4K91 mutants share the same mechanism (Figure 4A). Corroborating our findings from the developmental model, expression of H4K91R or H4K91Q caused upregulation of genes enriched in mature neuronal pathways, including synapse organization, neuron projection guidance and neuron migration (Figure 4B, Table S2), including known neuronal markers Map2, Tubb3, Rbfox3, and Dcx. (Figure 4C). Collectively, we demonstrate that H4K91 mutants alter gene expression and impact neural differentiation.
We next investigated the nuclear localization and the chromatin occupancy of H4K91 mutants in differentiated neural cells. Similar to mESCs (Figure 1A, F, Figure S1D–E) and neural cells in the brain (Figure 3B), staining with anti-HA antibody identified the distinct puncta staining pattern in differentiated neural cells that express H4 mutants, but not WT H4 (Figure 4D). This implies that H4K91 mutants share the ectopic recruitment pattern in distinct cell lineages. We further performed anti-HA ChIP-seq to characterize the genome-wide incorporation of H4 mutants. Previous studies have shown that most co-enrichment of H3.3 and H3K9me3 in mESCs is lost upon differentiation to NPCs, except at telomeres32,33. Similarly, we saw enhanced chromatin occupancy of H4K91 mutants at telomeric regions in NPCs or differentiated neural cells (Figure S4C). Additionally, H3.3 and H3K9me3 have previously been shown to localize at imprinted genes, which are specialized genes that are expressed in a monoallelic fashion34. We focused on 20 known imprinted loci that were shown to carry H3K9me341. We found the enrichment of H4K91 mutants at these imprinted loci in both mESCs and differentiated cells (Figure S4D). Interestingly, 12 out of 26 genes within these imprinted loci with enhanced occupancy of H4 mutants were significantly upregulated in mutant expressing neural cells (p<0.05, Figure S4D), as exemplified by select gene loci (Figure 4E). Taken together, the distinct puncta staining, combined with the increased enrichment of H4K91 mutants at telomeres and imprinted genes, suggest that the same co-localization with H3.3 and H3K9me3 identified in mESCs is also occurring in differentiated neural cells.
Discussion
Our study details how dominant histone H4 mutants at lysine 91 act through abnormal genomic localization, which to our knowledge demonstrates a new molecular pathogenic mechanism compared to previous research that focused on histone PTMs. We show that histone H4K91 mutants found in developmental disorders are incorporated into chromatin and preferentially associate with the histone variant H3.3, a histone variant that also has mutations in developmental disorders. Mediated by the H3.3-specific chaperone DAXX, the H3.3/H4K91 mutant complex is deposited into H3K9me3-enriched heterochromatin and forms distinct puncta in the nucleus of several distinct cell lineages (Figure S5). Expression of H4K91 mutant drives increased chromatin accessibility, consistent with our previous finding that H4 lysine 91 is critical for nucleosome stability in vitro21. Moreover, the accumulation of H4K91 mutants alters gene expression and promotes precocious neural differentiation, consistent with the reduced brain size and altered cortical layers observed in our mouse model (Figure S5). These findings recapitulate the microcephaly phenotype of patients carrying these germline mutations and are in line with the previously reported underdeveloped brain phenotype in H4K91 mutant zebrafish15. Our model will be instrumental in future efforts to elucidate the brain defects at distinct developmental stages, the cell-type specific genomic incorporation of H4K91 mutants into chromatin, as well as changes in chromatin accessibility and gene expression in post-mitotic neurons in vivo at distinct development times. These studies will clarify how H4K91 mutants lead to brain defects during development.
Like many histone lysine residues, H4K91 is subject to several post-translational modifications, including acetylation, mono-ubiquitination, and glutarylation35,42,43. A primary future direction will be to characterize the genome-wide distribution of H4K91 modifications in mutant expressing cells. So far, three different missense mutations have been identified in patients: H4K91E, H4K91Q, and H4K91R, which mimic glutarylated, acetylated, and unmodified lysine, respectively. All patients share similar clinical features, such as microcephaly and delayed development, which suggests that three mutated residues (E/R/Q) of H4K91 share a common mechanism of action in driving developmental phenotype. We propose that despite mimicking different post-translational modifications, these three documented H4K91 mutants converge to affect chromatin assembly and structure through nucleosome destabilization, altering basic cellular functions, such as DNA damage response and gene transcription, during development.
Besides histone H4K91, several de novo missense germline mutations affecting multiple residues of histone variant H3.3 have been reported in patients with progressive neurologic dysfunction and congenital anomalies17–20. A recent study of H3.3G34R substitution in mice found that the G34R mutant led to progressive microcephaly and neurodegeneration through impairing recruitment of the DNA methyltransferase DNMT3A and its redistribution on chromatin20. This study provides insights into the impact of H3.3 mutants on the chromatin landscape and highlights the roles of H3.3WT in regulating brain function. Furthermore, our study demonstrates that there is an enhanced association of H4K91 mutants with H3.3 and genome-wide colocalization of H4K91 mutants with H3.3. This study provides an intriguing molecular link between H4K91 mutants and the histone variant H3.3. We propose that H4K91 mutants and some H3.3 mutations may be incorporated into similar genomic regions, disrupting the local chromatin structure and altering gene expression. Further studies using H4K91 mutant mice and H3.3 conditional KO mice may help clarify the underlying regulatory mechanisms in the physiologic context of development. In light of the increasing number of histone germline mutations and the lack of understanding of their impact, our study here provides interesting biochemical, cellular, and molecular evidence for how mutations of H4 impact its binding properties with histone H3, altering H4 genomic incorporation, as well as derailing gene expression and cell fate during neural differentiation.
In addition to newly identified histone mutants, disturbances of imprinted gene expression due to genetic and epigenetic defects account for imprinting disorders, a group of congenital diseases affecting growth and development44. The enrichment of H4K91 mutants at imprinted loci and upregulation of several imprinted genes with H4K91 mutant expression suggests that mutated H4 might cause developmental defects through the aberrant expression of imprinted genes, which is one focus of our future studies. Together, our findings shed light on the effects of development by histone mutants beyond their well-known oncogenic roles.
Methods:
ESC culture and differentiation
ESCs are cultured in gelatin-coated plates and serum/LIF medium, which is composed of KO DMEM, 15% ES FBS, β-mercaptoethanol, Glutamax, LIF, Pen/Strep, and non-essential amino acid. To generate ESCs expressing HA-tagged H4 transgenes, H4WT or mutated H4 (H4K91E/R/Q) with 3-HA tagged at the c-terminal were cloned to PiggyBAC transposon plasmid. With the mouse ES Cell Nucleofector Kit (Lonza VPH-1001), PiggyBAC and pBASE plasmid are co-transfected into mESCs, which went through 2 weeks of G418 selection to isolate cells stably expressing H4 transgenes. H3.3 DKO, DAXX KO, and Hira KO mESCs were established previously31,45,46. To generate mESCs that transiently express GFP tagged H4 for live cell imaging or staining, H4WT or mutated H4 (H4K91E/R/Q) were cloned to the Xlone-GFP plasmid (addgene 96930), followed by the transfection with Xfect mESC transfection reagent (Takara 631317).
Neural differentiation protocol was modified from the previous method47. Briefly, mESCs were cultured with mESC medium in a gelatin-coated dish with MEF feeder cells for two passages, followed by culture without feeder cells for two or three passages. Then 2 million mESCs were plated to LIF-deprived medium and low-attached dishes (Greiner Bio One 633102) for 8 days to form embryonic body (EB) aggregates and 5uM retinoic acid (RA, Millipore R2625) were added to the medium from day 4. The medium was changed every two days. At day 8, EBs were washed and dissociated with fresh Trypsin_EDTA 0.05% (Gibco 25300054) to neuronal progenitor cells (NPCs). NPCs were resuspended in neurobasal medium, which includes neurocult basal medium plus proliferation supplement (stem cell Technologies, 05702), pen/strep, glutamax, EGF (Shenandoah Biotech #100–26) + FGF (Shenandoah Biotech #100–146). Then NPCs were plated on Poly_D_lysine (PDL, Millipore A-003-E) and Mouse laminin (sigma L2020–1MG) coated dishes. The neurobasal medium was changed 2h and one day after plating. Two days after plating, neural differentiation was initiated by switching to the neural differentiation medium (stem cell Technologies, 05704). After partially removing the old medium, a new differentiation medium was added every other day. Cell pellets were collected four days after differentiation for RNA isolation.
Immunostaining, in situ hybridization, ATAC-see, and Histology
mESCs, NPCs, or differentiated cells were plated on coated glass coverslips (Neuvitro CG-18-PDL). Cells were fixed with 1% fresh paraformaldehyde in PBS for 15–20 minutes and then washed with PBST (0.1% Triton). Then cells were blocked in PBST with 1% BSA (w/v) for 30 minutes before incubating with primary antibodies overnight at 4 °C, followed by secondary antibody staining at room temperature (RT) for 1h.
mESCs-derived mice at p0 were fixed with 2% fresh paraformaldehyde. After dissection, brain tissues were embedded in parafilm and prepared as 5μm sections by HistoWiz, Inc. To obtain similar cerebral cortex regions among different brain samples, sections were monitored by H&E staining. After rehydrating, brain sections were boiled in sodium citrate buffer (10mM sodium citrate, 0.05% Tween 20, pH 6.0) for 18 min, and then stained with diluted primary antibodies overnight at 4 °C and then secondary antibody at RT for 1h.
In situ hybridization (ISH) with the telomere probe Tamra-TelG (Tam-OO-TTAGGGTTAGGGTTAGGG 3’, synthesized from BioSynthesis, a gift from Dr. de Lange) was done following de Lange lab published protocols (https://delangelab.org/protocols). Briefly, cells on coverslips were fixed for 5 minutes at RT in 2% paraformaldehyde again. After a wash in PBS, cells were dehydrated in ethanol, consecutively 70%, 95%, and 100% EtOH for 5 minutes each. Next, dry the coverslip and apply a drop of the hybridizing solution including a diluted telomeric probe (1:3000 dilution). Then coverslips with the hybridizing solution were incubated for 3–10 minutes at 70–80 °C on heatblock, followed by incubation at RT in a humidity-controlled environment.
ISH with brain sections were done using commercially available RNA probes: Tbr2 (ACD, #429641-C2) and Satb2 (ACD, 413261-C3), following their user manual of RNAscope Multiplex Fluorescent Reagent Kit v2 assay.
ATAC-see in 293T cells was done with the EZ-Tn5™ Transposase (Lucigen TNP9211), following the published protocol36. Panobinostat (Cayman Chemicals 13280) was used to treat the 293T cells at the concentration of 200nM for 17h.
Imaging of all cells or tissues was done with a 40X or 63X objective and tile scan of Zeiss LSM 780 confocal microscope. Imaging analysis was performed using Zeiss Zen or image J software. Quantification of mutant puncta and ATAC-see puncta were done following the previous published protocol48. Following antibodies and dilutions were used: HA (Biolegend 901501, 1:400), ATRX (Santa Cruz, sc-15408, 1:200), DAXX (Santa Cruz, sc-8043, 1:200), Goat anti-Rabbit Alexa Fluor 488/568/647 (Invitrogen, 1:1000), DAPI (1:1000).
Immunoblot analysis
Cell pellets were resuspended in 1X Laemmli Sample Buffer and boiled for 10–15 minutes at 95°C. Samples were loaded to Tris-Glycine SDS-PAGE gels (16% or 4–20% gels, Invitrogen) and transferred to a nitrocellulose membrane. The blots were first stained with direct blue to detect all proteins, blocked with 1% milk in 1X TBST, and then incubated with diluted primary antibodies overnight at 4°C. After washing with TBST, secondary antibodies were then applied to blots for 1h at RT. Stained immunoblots were incubated in immobilon ECL solution (Millipore) and imaged using an Amersham Imager 600 (GE). Following antibodies were used: HA (Biolegend 901501, 1:1000), H3 (Abcam 1791, 1:10,000), H3.3 (Millipore 09–838, 1:1000), H4 (Abcam ab10158, 1:5000).
Mononucleosome immunoprecipitation (Mono-IP)
3HA-tagged H4WT, H4K91R, or H4K91Q were cloned into pCDH-EF1-MCS-Puro lentiviral vectors. HEK293T cells that stably express HA tagged H4WT or H4K91 mutants were generated as previously described10. HEK293T cells were collected from two 100% confluent 15cm dishes for each immunoprecipitation. Cells were washed with PBS and lysed on ice for 5 minutes in the following buffer: 10 mM HEPES pH 7.9, 10 mM KCl, 1.5 mM MgCl2, 0.34 M sucrose, 10% glycerol, 1mM DTT, protease inhibitor cocktail and Triton X-100 to 0.1%. Then cells were centrifuged at 3.5 K and 4°C for 5 minutes to collect nuclei. Nuclei pellets were washed in lysis buffer without Triton and then resuspended in no-salt solution (3 mM EDTA, 0.2 mM EGTA, 1 mM DTT, and 1× complete protease inhibitor cocktail) on ice with occasional vertexing to break the nuclei. Then microcentrifuge at 4K and 4°C for 5 minutes was done to collect chromatin fraction. Next chromatin pellet was resuspended in MNase digestion buffer (50mM HEPES, 2mM CaCl2, 0.2% NP-40, and 1× complete protease inhibitor cocktail). 18ul MNase (Worthington) was added to digest chromatin at 37°C for 10 minutes and the reaction was stopped by adding EDTA with the final concentration at 5mM. The digested chromatin was centrifuged at max speed for 10 minutes and the supernatant was collected. Take 5% of the supernatant and DNA was extracted by phenol-chloroform after RNase and Proteinase K treatment at 65°C for 1h. The fragment size of DNA was analyzed by 2% agarose gel electrophoresis to see if most nucleosomes were digested into mononucleosomes.
For the next step, digested mononucleosome was dialyzed with M.W. 3500 membrane (Thermo fisher 66330) against the dialysis buffer (20 mM HEPES pH 7.9, 20% glycerol, 0.2 mM EDTA, 0.2% TritonX-100, freshly added protease inhibitors) with 150mM KCl for 2h at 4°C. Then dialyzed mononucleosome was incubated with 100 ul anti-HA magnetic beads (Thermo 88837) overnight at 4°C while rotating. Then beads were washed three times using dialysis buffer with 150mM KCl and then three times through dialysis buffer with 100mM NaCl and 10% glycerol. Elution was done using a 5% acetic acid solution. Three replicates of monoIP pulldown were collected and analyzed by mass spectrometry analysis and immunoblot analysis.
Mass spectrometry
MonoIPs were dried in a centrifuge vacuum concentrator and resuspended in 20 ul of 50mM Ammonium Bicarbonate. Samples were propionylation as previously described49. In short, 10ul of propionic anhydride was added to 30ul of Acetonitrile and 10ul of the mixture was added to each sample and well mixed. Immediately, the pH of each sample was adjusted by addition of ammonium hydroxide to pH 8.0. Samples were incubated for 15 min at room temperature, dried down, and the propionylation was repeated. Samples were resuspended in 50mM Ammonium Bicarbonate and digested overnight with 2 ul of trypsin at room temperature. Digested peptides were dried down and the propionylation steps were repeated. Samples were desalted with self-packed C18 stage-tips and resuspended in 20ul 0.1% formic acid. The samples (2ul) were injected onto the Acclaim PepMap 100 C18 column (3 μm × 0.075 mm × 150 mm) for the analysis on a Q-Exactive Plus instrument (Thermo Fisher Scientific) attached to an UltiMate 3000 UHPLC (Thermo Fisher Scientific) and Nanospray Flex ion source (Thermo Fisher Scientific). The peptides were separated using buffer A (0.1% formic acid) and buffer B (80% acetonitrile and 0.1% formic acid) with a gradient of 2–35% over 50 min. The column was then washed at 98% buffer B over 5 min and equilibrated to 2% buffer B. Data-independent acquisition (DIA) was performed with the following settings: A full MS1 scan from 300 to 1100 m/z with a resolution of 70,000, an automatic gain control (AGC) target of 1 × 106, and a maximum injection time of 50 ms. DIA scans were attained across the same mass range with sequential isolation windows of 24 m/z with a normalized collision energy of 30, a resolution of 17,500, an AGC target of 2 × 105, and a maximum injection time of 60 ms. Data was analyzed with in-house program, EpiProfile.
Chromatin immunoprecipitation (ChIP)
For each immunoprecipitation, mESCs from two confluent T75 flasks were fixed with 1% paraformaldehyde for 5 minutes with gentle rotation and then quenched with 125 mM glycine for 5 minutes. To get chromatin, fixed cells were resuspended in 1ml of buffer 1 (50 mmol/L HEPES pH 7.5, 140 mmol/L NaCl, 1 mmol/L EDTA, 10% glycerol, 0.5% NP-40, 0.25% Triton X-100, and 1× complete protease inhibitor cocktail) and incubated for 10 minutes at 4°C with rotation. Samples were centrifuged at 1400× g for 5 minutes and pellets were resuspended in 1ml of buffer 2 (10 mmol/L Tris-HCl pH 8.0, 200 mmol/L NaCl, 1 mmol/L EDTA, 0.5 mmol/L EGTA, and 1× complete protease inhibitor cocktail) for incubation at 4°C for 10 minutes with rotation. Next samples were centrifuged at 1400×g for 5 minutes and resuspended in 900ul of buffer 3 (10 mmol/L Tris-HCl pH 8.0, 100mmol/L NaCl, 1 mmol/L EDTA, 0.5 mmol/L EGTA, 0.1% sodium deoxycholate, 0.5% N-Lauroylsarcosine, and 1× complete protease inhibitor cocktail). Samples were homogenized by passing through a syringe (28G1/2) eight times. Finally, 100ul of 10% Triton X-100 was added to homogenized chromatin for 16 minutes of sonication using a Covaris E220.
Sonicated samples were centrifuged for 10 minutes at 4°C. 5% of the supernatant was saved to extract DNA as input, while the rest was incubated with 75ul anti-HA magnetic beads (Thermo 88837) overnight at 4°C while rotating. Beads were washed six times with 1ml of cold RIPA buffer (50 mmol/L HEPES pH 7.5, 500 mmol/L LiCl, 1 mmol/L EDTA, 0.7% sodium deoxycholate, and 1% NP-40) and once with 1ml of wash buffer (10 mmol/L Tris-HCl pH 8.0, 50 mmol/L NaCl, and 1 mmol/L EDTA). Then beads were incubated with 210 ul of elution buffer (50 mmol/L Tris-HCl pH 8.0, 10 mmol/L EDTA, and 1% SDS) at 65°C for 3 minutes while shaking. The eluted chromatin was centrifuged at maximum speed for 1 minute at RT. Cross-linking was reversed by incubating the supernatant overnight at 65°C, followed by 1h RNaseA treatment at 37°C and 2h Proteinase K treatment at 55°C. DNA was extracted after mixing with phenol-chloroform and then ethanol precipitation.
For anti-HA ChIP with NPCs or differentiated neural cells, ten million cells were collected and fixed with 1% paraformaldehyde for 5 minutes with gentle rotation and then quenched with 125 mM glycine for 5 minutes. To get chromatin, fixed cells were washed with cold 1X PBS and lysed using previously published NEXSON protocol50. Briefly, cells were resuspended in 500ul of lysis buffer (5 mM PIPES, pH 8, 85 mM KCl, 0.5% NP-40, 1X complete Protease inhibitor cocktail) and sonicated in Bioruptor Plus (low power, 9 cycles of 15 seconds on and 30 seconds off). A small fraction of sonicated samples were checked under the microscope in between to monitor the lysis efficiency until more than 70% of cells were lysed. Then lysed samples were centrifuged for 5 minutes at 1000×g at 4 °C. Nuclei pellets were washed in lysis buffer and resuspended in 130ul sonication buffer (10 mM Tris-HCl, pH 8, 0.1% SDS, 1 mM EDTA, 1X Complete Protease inhibitor cocktail) for 6min sonication using a Covaris E220. The sheared chromatin was incubated with 4ul HA antibody (Biolegend 901501, 1:1000) and immunoprecipitation was done using ChIP-IT high-sensitive kit (active motif 53040).
ChIP libraries were generated with input and pulldown DNA using NEBNext Ultra II DNA Library Prep Kit for Illumina (NEB E7645L) for sequencing on an Illumina Nextseq with 75-bp read length and single-end.
RNA isolation and sequencing
Total RNA was isolated from cells using the RNeasy kit (Qiagen) and analyzed using the Bioanalyzer RNA Pico (Agilent) before library preparation. PolyA mRNA enrichment was performed using the NEB Next Poly(A) mRNA magnetic isolation module (NEB E7490L), followed by library preparation using the NEB Next Ultra II RNA Library Prep Kit (NEB E7770L). Sequencing was done using the Illumina Nextseq with 75-bp read length and single-end.
Bioinformatic analysis
RNA-seq analysis
Transcript abundance was determined from FASTQ files using Salmon (v0.8.1) and the GENCODE reference transcript sequences51. Transcript counts were imported into R with the tximport R Bioconductor package (v1.8.0), and differential gene expression was performed with the DESeq2 R Bioconductor package (v1.20.0)52,53. Normalized counts were retrieved from the DESeq2 results and z-scores for the indicated gene sets were visualized with heatmaps generated using the pheatmap R package (v1.0.12)54. Gene ontology analysis was performed with the selected gene lists using the clusterProfiler R Bioconductor package (v4.0.5)55.
ChIP-seq analysis
ChIP-seq reads were aligned using the Rsubread R Bioconductor package (v1.30.6) and predicted fragment lengths were calculated by the ChIPQC R Bioconductor package (v1.16.2)56,57. Normalized, fragment-extended signal bigWigs were created using the rtracklayer R Bioconductor package (v1.40.6), and peaks were called using MACS2 (v2.1.1)58,59. Range-based heatmaps showing signal over genomic regions were generated using the profileplyr R Bioconductor package (v1.8.1)60. To identify K91mut enriched peaks, the union of HA peaks from H4WT, H4K91R, and H4K91Q cells was filtered to those with a p value less than 10^−10 (as reported from MACS2), and then peaks from empty vector cells were removed. K-means clustering was used on the regions around these peaks to cluster the signal heatmaps and identify a cluster with high signal in both H4 mutant samples and low signal in H4WT expressing cells. Peaks were annotated with the various types of genomic regions using the ChIPseeker R Bioconductor package (v1.28.3)61. Any regions included in the ENCODE blacklisted regions of the genome were excluded from all region-specific analyses62. To assess ChIP-seq signal in telomeres, fastq files were aligned to a DNA sequence of 150 conserved telomere repeats (TTAGGG) using the R Bioconductor package Rbowtie2 (v1.12.0)63,64.
Genome-wide correlation analysis
The signal from ChIP samples was quantified in 20bp bins +/− 1kb from the center of the HA union peak set described above. These bins were then ranked by the indicated ChIP sample, and the bins were then divided into 100 quantiles for the x-axis of the correlation plot. The signal of all ChIPs in these ranked quantiles was then plotted using the geom_smooth() function from the ggplot2 R package (v3.3.6)65.
Tetraploid complementation and all-ESC mutant mice
Animals were housed and cared for according to a protocol approved by the IACUC of Weill Cornell Medical College (Protocol number: 2014–0061). Wild-type ICR mice were purchased from Taconic Farms (Germantown, NY). Females were super-ovulated at 6–8 weeks with 0.1 ml CARD HyperOva (Cosmo Bio Co., Cat. No. KYD-010-EX) and 5 IU hCG (Human chorionic gonadotrophin, Sigma-Aldrich) at intervals of 48 hours. The females were mated individually to males and checked for the presence of a vaginal plug the following morning. Plugged females were sacrificed at 1.5 days post-hCG injection to collect 2-cell embryos. Embryos were flushed from the oviducts with advanced KSOM (Cat # MR-101-D, Millipore). The 2-cell embryos were subjected to electrofusion to induce tetraploidy. Fused embryos were moved to new KSOM micro drops covered with mineral oil and cultured to the blastocyst stage until ESC injection.
ESCs expressing HA-tagged H4WT, H4K91R, and H4K91Q were trypsinized, resuspended in ESC medium, and kept on ice. A flat tip microinjection pipette was used for ESC injections. ESCs were collected at the end of the injection pipette and 10–15 cells were injected into each 4n blastocyst. The injected 4n blastocysts were kept in KSOM until embryo transfer. Typically, ten injected 4n blastocysts were transferred into each uterine horn of 2.5 dpc pseudo-pregnant ICR females. After injecting ESCs into tetraploid blastocysts, the 4n cells of the host embryo contribute solely to the placenta, while the injected ESCs form the embryo properly. All-ESC pups were recovered via cesarean section at embryonic day 19.5 (E19.5), which is equivalent to postnatal day 0 (P0) in normal fertilized embryos Mice genotyping was done using PCR with the following two primers:
Forward: 5’ CGGACTAGTGCCACCATGTCTGG 3’
Reverse: 5’ AAATATGCGGCCGCTTAAGCATAATCAGGCACAT 3’
Supplementary Material
Acknowledgments:
We thank past and present members of the Allis laboratory, especially Leah Gates, Agata Lemiesz Patriotis, and Marylene Leboeuf. We acknowledge The Rockefeller University for financial support and the community, especially Shixin Liu, Viviana Risca, Hironori Funabiki, and Michael W. Young for helpful discussions and support. We thank Shasha Chong for sharing the ImageJ algorithm used to quantify the puncta intensity. We thank Francisca Vitorino from Garcia lab for uploading the Mass Spectrometry data to the MassIVE database. Finally, we acknowledge the help from the following resource centers at The Rockefeller University: Bio-Imaging, Bioinformatics, and Genomics. This work was supported by the NIH grant P01CA196539 to C.D.A. L.F. was supported by the C. H. Li Memorial Scholar Fund at The Rockefeller University and an NIH Career Award (K99HD107908). E.G.P. and B.A.G. were supported by NIH grants R01NS111997 and R01HD106051. L.Z. and D.W. were supported by the NIH grant (R01GM129380 to D.W.). A.A.S. is supported by UTSA and NCI grant R01CA234561. This article is dedicated to the memory of C. David Allis, who passed away on January 8, 2023.
Footnotes
Declaration of Interests
The authors declare no competing interests.
Data availability
The ChIP-seq and RNA-seq data have been deposited to the Gene Expression Omnibus (GEO) database under accession number GSE231567. The mass spectrometry data has been deposited to the MassIVE database under dataset number MSV000091837. Any additional information or materials are available upon request.
References
- 1.Bjornsson H. T. The Mendelian disorders of the epigenetic machinery. Genome Research vol. 25 Preprint at 10.1101/gr.190629.115 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Flavahan W. A., Gaskell E. & Bernstein B. E. Epigenetic plasticity and the hallmarks of cancer. Science vol. 357 Preprint at 10.1126/science.aal2380 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Behjati S. et al. Distinct H3F3A and H3F3B driver mutations define chondroblastoma and giant cell tumor of bone. Nat Genet 45, 1479–1482 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Khuong-Quang D.-A. et al. K27M mutation in histone H3.3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta Neuropathol 124, 439–447 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Schwartzentruber J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012). [DOI] [PubMed] [Google Scholar]
- 6.Wu G. et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet 44, 251–253 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Nacev B. A. et al. The expanding landscape of ‘oncohistone’ mutations in human cancers. Nature 567, 473–478 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Arimura Y. et al. Cancer-associated mutations of histones H2B, H3.1 and H2A.Z.1 affect the structure and stability of the nucleosome. Nucleic Acids Res 46, 10007–10018 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bennett R. L. et al. A Mutation in Histone H2B Represents a New Class of Oncogenic Driver. Cancer Discov 9, 1438–1451 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lewis P. W. et al. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340, 857–61 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lu C. et al. Histone H3K36 mutations promote sarcomagenesis through altered histone methylation landscape. Science (1979) 352, 844–849 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chan K. M. et al. The histone H3.3K27M mutation in pediatric glioma reprograms H3K27 methylation and gene expression. Genes Dev 27, 985–990 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Fang D. et al. The histone H3.3K36M mutation reprograms the epigenome of chondroblastomas. Science (1979) 352, 1344–1348 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tessadori F. et al. A de novo variant in the human HIST1H4J gene causes a syndrome analogous to the HIST1H4C -associated neurodevelopmental disorder. European Journal of Human Genetics (2019) doi: 10.1038/s41431-019-0552-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tessadori F. et al. Germline mutations affecting the histone H4 core cause a developmental syndrome by altering DNA damage response and cell cycle control. Nat Genet 49, 1642–1646 (2017). [DOI] [PubMed] [Google Scholar]
- 16.Tessadori F. et al. Recurrent de novo missense variants across multiple histone H4 genes underlie a neurodevelopmental syndrome. Am J Hum Genet 109, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bryant L. et al. Histone H3.3 beyond cancer: Germline mutations in Histone 3 Family 3A and 3B cause a previously unidentified neurodegenerative disorder in 46 patients. Sci Adv 6, 1–12 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Maver A., Čuturilo G., Ruml S. J. & Peterlin B. Clinical next generation sequencing reveals an H3F3A gene as a new potential gene candidate for microcephaly associated with severe developmental delay, intellectual disability and growth retardation. Balkan Journal of Medical Genetics 22, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Okur V. et al. De novo variants in H3–3A and H3–3B are associated with neurodevelopmental delay, dysmorphic features, and structural brain abnormalities. NPJ Genom Med 6, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Khazaei S. et al. Single substitution in H3.3G34 alters DNMT3A recruitment to cause progressive neurodegeneration. Cell 186, 1162–1178.e20 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bagert J. D. et al. Oncohistone mutations enhance chromatin remodeling and alter cell fates. Nat Chem Biol 17, 403–411 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bender S. et al. Reduced H3K27me3 and DNA Hypomethylation Are Major Drivers of Gene Expression in K27M Mutant Pediatric High-Grade Gliomas. CCELL 24, 660–672 (2013). [DOI] [PubMed] [Google Scholar]
- 23.Irgen-Gioro S., Yoshida S., Walling V. & Chong S. Fixation can change the appearance of phase separation in living cells. Elife 11, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Elsässer S. J. et al. DAXX envelops a histone H3.3-H4 dimer for H3.3-specific recognition. Nature 491, 560–565 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lewis P. W., Elsaesser S. J., Noh K. M., Stadler S. C. & Allis C. D. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci U S A 107, 14075–14080 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Drané P., Ouararhni K., Depaux A., Shuaib M. & Hamiche A. The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev 24, 1253–1265 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ray-Gallet D. et al. HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol Cell 9, (2002). [DOI] [PubMed] [Google Scholar]
- 28.Smith S. & Stillman B. Purification and characterization of CAF-I, a human cell factor required for chromatin assembly during DNA replication in vitro. Cell 58, (1989). [DOI] [PubMed] [Google Scholar]
- 29.Wong L. H. et al. Histone H3.3 incorporation provides a unique and functionally essential telomeric chromatin in embryonic stem cells. Genome Res 19, 404–414 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wong L. H. et al. ATRX interacts with H3.3 in maintaining telomere structural integrity in pluripotent embryonic stem cells. Genome Res 20, 351–360 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Goldberg A. D. et al. Distinct Factors Control Histone Variant H3.3 Localization at Specific Genomic Regions. Cell 140, 678–691 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Navarro C., Lyu J., Katsori A. M., Caridha R. & Elsässer S. J. An embryonic stem cell-specific heterochromatin state promotes core histone exchange in the absence of DNA accessibility. Nat Commun 11, 1–14 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Elsässer S. J., Noh K. M., Diaz N., Allis C. D. & Banaszynski L. A. Histone H3.3 is required for endogenous retroviral element silencing in embryonic stem cells. Nature 522, 240–244 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Voon H. P. J. et al. ATRX Plays a Key Role in Maintaining Silencing at Interstitial Heterochromatic Loci and Imprinted Genes. Cell Rep 11, 405–418 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ye J. et al. Histone H4 lysine 91 acetylation a core domain modification associated with chromatin assembly. Mol Cell 18, 123–30 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chen X. et al. ATAC-see reveals the accessible genome by transposase-mediated imaging and sequencing. Nat Methods 13, 1013–1020 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wen D., Saiz N., Rosenwaks Z., Hadjantonakis A. & Rafii S. Completely ES Cell-Derived Mice Produced by Tetraploid Complementation Using Inner Cell Mass ( ICM ) Deficient Blastocysts. 9, (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tiberi L., Vanderhaeghen P. & van den Ameele J. Cortical neurogenesis and morphogens: Diversity of cues, sources and functions. Curr Opin Cell Biol 24, 269–276 (2012). [DOI] [PubMed] [Google Scholar]
- 39.Maze I. et al. Critical Role of Histone Turnover in Neuronal Transcription and Plasticity. Neuron 87, 77–94 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Xia W. & Jiao J. Histone variant H3.3 orchestrates neural stem cell differentiation in the developing brain. Cell Death Differ 24, 1548–1563 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mikkelsen T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature (2007) doi: 10.1038/nature06008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bao X. et al. Glutarylation of Histone H4 Lysine 91 Regulates Chromatin Dynamics Article Glutarylation of Histone H4 Lysine 91 Regulates Chromatin Dynamics. Mol Cell 1–16 (2019) doi: 10.1016/j.molcel.2019.08.018. [DOI] [PubMed] [Google Scholar]
- 43.Yan Q. et al. Article BBAP Monoubiquitylates Histone H4 at Lysine 91 and Selectively Modulates the DNA Damage Response. Mol Cell 36, 110–120 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Monk D., Mackay D. J. G., Eggermann T., Maher E. R. & Riccio A. Genomic imprinting disorders: lessons on how genome, epigenome and environment interact. Nature Reviews Genetics Preprint at 10.1038/s41576-018-0092-0 (2019). [DOI] [PubMed] [Google Scholar]
- 45.Roberts C. et al. Targeted Mutagenesis of the Hira Gene Results in Gastrulation Defects and Patterning Abnormalities of Mesoendodermal Derivatives Prior to Early Embryonic Lethality . Mol Cell Biol 22, (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Banaszynski L. A. et al. Facilitates PRC2 Recruitment at Developmental Loci in ES Cells. 5, 107–120 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bibel M., Richter J., Lacroix E. & Barde Y. A. Generation of a defined and uniform population of CNS progenitors and neurons from mouse embryonic stem cells. Nat Protoc 2, 1034–1043 (2007). [DOI] [PubMed] [Google Scholar]
- 48.Wan L. et al. Impaired cell fate through gain-of-function mutations in a chromatin reader. Nature 577, 121–126 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sidoli S., Bhanu N. V., Karch K. R., Wang X. & Garcia B. A. Complete workflow for analysis of histone post-translational modifications using bottom-up mass spectrometry: From histone extraction to data analysis. Journal of Visualized Experiments 2016, (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Arrigoni L. et al. Standardizing chromatin research: A simple and universal method for ChIP-seq. Nucleic Acids Res 44, (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Patro R., Duggal G., Love M. I., Irizarry R. A. & Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods 14, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Soneson C., Love M. I. & Robinson M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Love M. I., Huber W. & Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kolde R. pheatmap : Pretty Heatmaps. R package version 1.0.8 (2015). [Google Scholar]
- 55.Yu G., Wang L. G., Han Y. & He Q. Y. ClusterProfiler: An R package for comparing biological themes among gene clusters. OMICS 16, (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Carroll T. S., Liang Z., Salama R., Stark R. & de Santiago I. Impact of artifact removal on ChIP quality metrics in ChIP-seq and ChIP-exo data. Front Genet 5, (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Liao Y., Smyth G. K. & Shi W. The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads. Nucleic Acids Res 47, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lawrence M., Gentleman R. & Carey V. rtracklayer: An R package for interfacing with genome browsers. Bioinformatics 25, (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhang Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol 9, (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Barrows D. and profileplyr C. T.: Visualization and annotation of read signal over genomic ranges with profileplyr. Bioconductor (2019). [Google Scholar]
- 61.Yu G., Wang L. G. & He Q. Y. ChIP seeker: An R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, (2015). [DOI] [PubMed] [Google Scholar]
- 62.Amemiya H. M., Kundaje A. & Boyle A. P. The ENCODE Blacklist: Identification of Problematic Regions of the Genome. Sci Rep 9, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Au K. F., Jiang H., Lin L., Xing Y. & Wong W. H. Detection of splice junctions from paired-end RNA-seq data by SpliceMap. Nucleic Acids Res 38, (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Langmead B., Trapnell C., Pop M. & Salzberg S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10, (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wickham H. ggplot2: Elegant Graphics for Data Analysis. Springer-Verlag; New York. Media vol. 35 (2016). [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The ChIP-seq and RNA-seq data have been deposited to the Gene Expression Omnibus (GEO) database under accession number GSE231567. The mass spectrometry data has been deposited to the MassIVE database under dataset number MSV000091837. Any additional information or materials are available upon request.