XB-ART-59531
Nat Commun
2023 Jan 28;141:472. doi: 10.1038/s41467-023-36114-x.
Show Gene links
Show Anatomy links
The H2A.Z and NuRD associated protein HMG20A controls early head and heart developmental transcription programs.
Herchenröther A
,
Gossen S
,
Friedrich T
,
Reim A
,
Daus N
,
Diegmüller F
,
Leers J
,
Sani HM
,
Gerstner S
,
Schwarz L
,
Stellmacher I
,
Szymkowiak LV
,
Nist A
,
Stiewe T
,
Borggrefe T
,
Mann M
,
Mackay JP
,
Bartkuhn M
,
Borchers A
,
Lan J
,
Hake SB
.
Abstract
Specialized chromatin-binding proteins are required for DNA-based processes during development. We recently established PWWP2A as a direct histone variant H2A.Z interactor involved in mitosis and craniofacial development. Here, we identify the H2A.Z/PWWP2A-associated protein HMG20A as part of several chromatin-modifying complexes, including NuRD, and show that it localizes to distinct genomic regulatory regions. Hmg20a depletion causes severe head and heart developmental defects in Xenopus laevis. Our data indicate that craniofacial malformations are caused by defects in neural crest cell (NCC) migration and cartilage formation. These developmental failures are phenocopied in Hmg20a-depleted mESCs, which show inefficient differentiation into NCCs and cardiomyocytes (CM). Consequently, loss of HMG20A, which marks open promoters and enhancers, results in chromatin accessibility changes and a striking deregulation of transcription programs involved in epithelial-mesenchymal transition (EMT) and differentiation processes. Collectively, our findings implicate HMG20A as part of the H2A.Z/PWWP2A/NuRD-axis and reveal it as a key modulator of intricate developmental transcription programs that guide the differentiation of NCCs and CMs.
PubMed ID: 36709316
PMC ID: PMC9884267
Article link: Nat Commun
Species referenced: Xenopus laevis
Genes referenced: bend3 brd2 cdh2 cdk2ap1 cdk2ap2 chd3 chd4 eif4h gata4 gatad2a gatad2b gse1 h2ac21 h2az1 h2az2 hdac1 hdac2 hmg20a hprt1 isyna1 itk l3mbtl3 mbd2 mbd3 mta1 mta2 myh6 phf14 phf21a pwwp2a rai1 rbbp4 rbbp7 rcor1 rcor3 rpe rpl11 tcf20 tead1 twist1 znf219 znf512b
GO keywords: chromatin binding [+]
chromatin organization
heart development
embryo development
embryonic neurocranium morphogenesis
cartilage development
Morpholinos: hmg20a MO1
Disease Ontology terms: autism spectrum disorder [+]
Phenotypes: Xla Wt + hmg20a MO (Fig 4.A r2c1-3) [+]
Xla Wt + hmg20a MO
(Fig. 4 B r1c4)
Xla Wt + hmg20a MO (Fig. 4 G r1c3)
Xla Wt + hmg20a MO (Fig. 4 G r2c3)
Xla Wt + hmg20a MO (Fig. 4E)
Xla Wt + hmg20a MO (Fig. 4 G r1c3)
Xla Wt + hmg20a MO (Fig. 4 G r2c3)
Xla Wt + hmg20a MO (Fig. 4E)
Article Images: [+] show captions
Fig. 1: HMG20A binds several chromatin-modifying complexes. A Immunoblots of GFP, HMG20A, BRD2 and PWWP2A (positive controls) as well as H3 upon GFP, GFP-H2A and GFP-H2A.Z.1 mononucleosome IPs. B Immunofluorescence microscopy images of GFP, GFP-HMG20A and endogenous HMG20A proteins (488 nm, green) in HeLaK cells. DNA is stained with Hoechst (blue). Scale bar: 20 µm. C Volcano plot of label-free interaction partners of GFP-HMG20A-associated mononucleosomes. Significantly enriched proteins over GFP-associated mononucleosomes are shown in the upper right part. t-Test (two-tailed) differences were obtained by two-sample t-test. HMG20A is highlighted in bright green, PRTH members in red, BHC/CoREST members in brown, NuRD members in blue, other proteins in black and background binding proteins in grey. See also Supplementary Fig. 1E for Volcano plot of second biological replicate and Supplementary Data 1 for detailed list of HMG20A binders. D Heatmap of significant outliers from two independent GFP-HMG20A mononucleosome IPs analysed by lf-qMS/MS (see C and Supplementary Fig. 1E) normalized to GFP. Scale bar: log2-fold t-test differences (two-tailed). E Immunoblots of GFP and GFP-HMG20A mononucleosome-IPs detecting endogenous members of the BHC/CoREST (HMG20B) and NuRD (MBD2, HDAC2) complexes as well as ZNF512B protein. Experiments in A, B, E were repeated independently three times with consistency. Source data for these figures are provided as a Source Data file. | |
Fig. 2: HMG20A binds NuRD complex components and DNA. A Anti-GFP immunoprecipitations of HEK293 cell extracts co-transfected with GFP-HMG20A and FLAG-HDAC1, -MTA1, -MTA2 and -RBBP4. Proteins were detected with anti-FLAG or anti-GFP antibodies. B Top: Anti-GFP immunoprecipitations of HEK293 cell extracts co-transfected with GFP-HMG20A and FLAG-CHD4 (CHD4), -CHD4-N-terminus (CHD4-N), -CHD4-middle domain (CHD4-M), -CHD4-C-terminus (CHD4-C) and HA-GATAD2A, -MBD2 and -MBD3. Proteins were detected with anti-FLAG and anti-HA or anti-GFP antibodies. Bottom: schematic depiction of CHD4 deletion constructs. C Top: schematic depiction of HMG20A deletion constructs. Bottom: anti-GFP IPs of HEK293 cell extracts co-transfected with GFP-HMG20A and its deletions (HMG, CC) and of NuRD members (FLAG-MTA1, HDAC1-FLAG, FLAG-RBBP4). Proteins were detected with anti-FLAG or anti-GFP antibodies. D EMSA of increasing amounts of extracts from Sf9 cells expressing FLAG-HMG20A and its deletions (see C, top) using Cy5-labelled DNA. Experiments in A-E were repeated independently at least two times with consistency. Source data for these figures are provided as a Source Data file. | |
Fig. 3: HMG20A localizes to distinct regulatory chromatin regions. A Genome browser snapshot of a representative region in human chromosome 15 displaying input (grey), GFP control (grey), H3K27ac, (purple), H3K4me3 (light green), H3K4me1 (dark green), GFP-H2A.Z.1 (red), GFP-PWWP2A (orange) and two replicates of GFP-HMG20A (blue) ChIP-seq signals (Pearson’s r = 0.88). Blue bar depicts HMG20A + H2A.Z.1 + PWWP2A-positive site, red bar depicts HMG20A-only site and green bar depicts a negative control site. B Venn diagram displaying numbers of HMG20A-bound sites and ENCODE published DNase I hypersensitive sites and their overlaps. C Enrichment plot representing genomic features of HMG20A + H2A.Z and HMG20A-only ChIP-seq sites. D Average binding profiles across transcriptional start sites of GFP-HMG20A (blue), -H2A.Z.1 (red), -PWWP2A (orange) and H3K4me3 (green) mean coverage signals at TSS of expressed genes. E ChromHMM33,34-based enrichment of chromatin states (defined by the specific combinatorial occurrence of five histone modifications) of GFP-HMG20A-only compared to HMG20A + H2A.Z-containing genomic regions. F Average binding plot of ENCODE H3K36me3-containing regions over HMG20A-only (yellow) and HMG20A + H2A.Z (blue) ChIP-seq sites. G Immunoblot of endogenous HMG20A upon siRNA-mediated depletion in HelaK cells (shown are three replicates). H Volcano plot of significantly deregulated (log2 fold change < −1, p < 0.05, calculated with Deseq2) mRNAs from two independent siRNA-mediated HMG20A depletion experiments analyzed by mRNA-seq. Red: upregulated transcripts, blue: downregulated transcripts. Source data for these figures are provided as a Source Data file. | |
Fig. 4: HMG20A depletion leads to craniofacial and heart malformations in frog. A Loss of function of Hmg20a leads to craniofacial and pigmentation defects in Xenopus tadpoles. *Marks the injected side, white arrow marks pigmentation defects. Scale bar = 1 mm. B Mean percentage of craniofacial defects of three independent experiments ± s.e.m. Number of embryos are indicated for each column. **p = 0.0024 (two-tailed unpaired Student’s t-test). C Hmg20 loss-of-function NC migration defects can partially be rescued by co-injection of human HMG20A DNA. * Marks the injected side (blue: lacZ staining, purple: twist staining), arrow indicates cranial NC migration defect. Scale bar = 1 mm. D NC migration defects of three independent experiments injected and analysed as shown in (C). Data are presented as mean ± s.e.m., **p (left) = 0.0026, ***p = 0.0002 ****p = 0.0001, **p (right) = 0.006 (one-way ANOVA, Tukey’s multiple comparisons test). E Hmg20a-depleted Xenopus tadpole embryos show defects in cartilage formation (arrow). For rescue experiments, human HMG20A DNA was co-injected, *marks the injected side. Scale bar = 500 µm. F Box and whiskers plots summarize cartilage defects of at least three independent experiments analysed as in (E). Number of embryos (n, above each bar) and median are indicated. The box extends from 25th to 75th percentile, with whiskers from minimum to maximum. **p = 0.0013, ****p = 0.0001, ns.: not significant (one-way ANOVA, Tukey’s multiple comparisons test). G Hmg20a loss-of-function causes heart defects. Top: mhcα in situ hybridization reveals defects in the formation of the first heart field (arrow) at stage 26. Bottom: At stage 42, the three-chambered heart structure consisting of two atria (a) and a ventricle (v) is disturbed; the malformed heart is displaced towards the injected side (arrow). The jaw muscle (jm), which is also marked by mhcα, is also reduced. Scale bar = 1 mm. H Graph summarizing three independent experiments as shown in (G), data are presented as mean ± s.e.m. *p = 0.0242, **p = 0.0038 (two-tailed unpaired Student’s t-test). Source data for these figures are provided as a Source Data file. | |
Fig. 5: HMG20A is essential for cardiomyocyte and neural crest differentiation in mESCs.A Validation of three Hmg20a DP clones by RT-qPCR. Shown is Hmg20a expression normalized to Hprt. Data is presented as mean ± SEM of three technical replicates. B Immunoblot analyses of endogenous HMG20A protein of extracts from WT and three Hmg20a DP mESC clones. H3 served as loading control (shown is one representative blot of two consistent replicates). C mESC neural crest cell (NCC) differentiation scheme. Created with BioRender.com. D RT-qPCR of neural crest and EMT marker genes in WT and three individual Hmg20a DP clones at Day9 of neural crest differentiation protocol normalized to Hprt, 18 S RNA and Gapdh expression. Data is presented as mean ± SEM of three technical replicates. E RT-qPCR of Hmg20a mRNA in WT and three individual Hmg20a DP clones at Day9 of neural crest differentiation protocol. Expression was normalized to Hprt, 18 S RNA and Gapdh expression. Data is presented as mean ± SEM of three technical replicates. F mESC cardiomyocyte (CM) differentiation scheme. Created with BioRender.com. G Depiction of percent beating (gray) or non-beating (black) WT or three individual Hmg20a DP EBs at Day7 (Top) or Day10 (bottom) of the cardiomyocyte differentiation procedure (see G). Ability to form contracting cardiomyocytes on Day7.5 is significantly reduced in all Hmg20a DP clones (Fischer’s exact test, two-sided, p = 3.1213974265e-087 (#06), p = 1.1876278705e-122 (#26), p = 2.0898552635e-091 (#48)). H RT-qPCR of Hmg20a mRNA in WT and three individual Hmg20a DP clones at Day7.5 of CM protocol. Expression was normalized to Hprt, 18 S RNA and Gapdh expression. Data is presented as mean ± SEM of three technical replicates. Source data for these figures are provided as a Source Data file. | |
Fig. 6: HMG20A regulates cardiomyocyte transcription programs. A Stacked Bar plot of numbers of significantly up (log2 fold change >1) and down (log2 fold change < −1) regulated genes (adjusted p-value < 0.05) during indicated time points of cardiomyocyte differentiation of WT and Hmg20a DP clone #26 mESCs as identified by mRNA-seq. B Principle component Analysis (PCA) of RNA-seq data of two replicates of WT (circle) and Hmg20a DP clone#26 (triangle) at Day0 (yellow), Day2 (magenta), Day4 (olive), Day6 (green) and Day7.5 (red) differentiation time points (see Fig. 5F for cardiomyocyte differentiation scheme). C Heat map showing the z-scaled expression values from all significant deregulated genes comparing the differentiation steps (Day 2 vs. 4; 4 vs. 6; 6 vs. 7.5). Genes are clustered according to the Euclidean distance by an unsupervised agglomerative hierarchical approach. Shown are the mean z-scales of two replicates for each day for WT (left panel) or Hmg20a DP (right panel) cells. D Line plots showing the min-max normalized mRNA-seq expression values for known cardiomyocyte marker genes at various differentiation time points. Plotted are mean expression values (dotted line) and the standard deviation (continuous line) for WT (blue) or Hmg20a DP (grey) cells. E RT-qPCR of cardiomyocyte marker genes in WT and three individual Hmg20a DP clones at Day7.5 of cardiomyocyte differentiation protocol normalized to Hprt, 18 S RNA and Gapdh expression. Data is presented as mean ± SEM of three technical replicates. Source data for these figures are provided as a Source Data file. | |
Fig. 7: HMG20A is enriched at open chromatin regions and affects chromatin accessibility in mESCs A Genome browser snapshots of representative HMG20A binding regions, as identified by CUT&RUN-seq of primed stage WT (blue) or Hmg20a DP clone #26 (light gray, negative control) mESCs. IgG antibody was used as negative control (black). Shown are two independent replicates (rep). B Venn diagram depicting overlay of CUT&RUN identified HMG20A binding sites with ATAC-seq identified accessible chromatin regions. C Density heatmap of ATAC-seq sites (two replicates) that become more open (cluster 1), remain unaffected (cluster 2) or become more closed (cluster 3) upon Hmg20a depletion. D Cumulative density plot showing the distribution of the observed changes in chromatin accessibility (shown as log2FC (DP/ET)) for all ATAC-seq signals (black) and for those ATAC-seq signals overlapping with HMG20A (red). DARs: differentially accessibly regions. | |
Fig. 9: Model of HMG20A’s function in chromatin and transcriptional regulation during development. Top: HMG20A associates with H2A.Z- and PWWP2A-associated PRTH and M1HR complexes and ZNF512B as well as BHC/CoREST and NuRD complexes and TEAD and L3MBTL3 that are not part of H2A.Z or PWWP2A interactomes. Middle: HMG20A binds to two distinct chromatin regulatory elements: (1) Nucleosome depleted regions (NDR) at promoter sites that are surrounded by H2A.Z-containing nucleosomes and bound by PWWP2A and that are associated with genes involved in basic processes, such as ‘chromatin organization‘. (2) H2A.Z-lacking intronic enhancers within transcribed genes belonging to developmental processes, such as ‘embryonic morphology”. Bottom: Depletion of HMG20A in Xenopus laevis and mESCs leads to changes in chromatin accessibility, deregulation of transcription programs as well as migration defects. HMG20A depleted cells fail to properly differentiate into neural crest cells or cardiomyocytes in mESCs as well as head and heart in Xenopus laevis. Figure was created with BioRender. | |
Supplementary Figure 1: HMG20A binds chromatin modifying complexes. (A) Schematic depiction of human HMG20A protein with its N-terminal HMG box and C-terminal coiled-coil (CC) domain. (B) Flow cytometry analysis of HeLaK cells (WT, brown) stably expressing GFP (green) or GFP-HMG20A (blue). (C) Immunoblot of cell extracts from HeLaK cells (WT) stably expressing GFP or GFP- HMG20A with anti-HMG20A antibodies. Anti-H3 serves as loading control. (D) Agarose gel of purified DNA fragments after MNase digestion using HeLaK cells stably expressing GFP and GFP-HMG20A. (E) Volcano plot of second replicate of label-free interaction partners of GFP-HMG20A- associated mononucleosomes. Significantly enriched proteins over GFP-associated mononucleosomes are shown in upper right part. T-test differences were obtained by two-sample t-test. HMG20A is highlighted in bright green, PRTH members in red, BHC/CoREST members in brown, NuRD members in blue, other proteins in black and background binding proteins in grey. See also Figure 1C for Volcano plot of first biological replicate and Supplemental Table 1 for detailed list of HMG20A binders. (F) Schematic depiction of overlapping H2A.Z.11,2, PWWP2A3 and HMG20A interactomes. Experiments in C, D were repeated independently three times with consistency. Source data for these figures are provided as a Source Data file. | |
Supplementary Figure 2: Expression and purification of HMG20A deletion proteins. (A) Alignment of HMG box amino acid sequences from diverse human HMG-box containing proteins. Alignment was performed using Clustal Omega. (B, C) Coomassie-stained SDS-PAGE gel (B) or anti-FLAG immunoblot (C) of extracts from Sf9 cells expressing FLAG-tagged HMG20A, HMG or CC proteins. Shown is one representative gel/blot of two consistent experiments. (D) Electrophoretic mobility shift assay (EMSA) of a Cy5-labeled DNA probe together with whole Sf9 extracts containing different FLAG-HMG20A proteins (see above) in increasing amounts. EMSA with whole cell extracts of uninfected Sf9 cells served as negative control. Source data for these figures are provided as a Source Data file. | |
Supplementary Figure 3: HMG20A binds regulatory genomic regions but is not correlated to transcriptional output in HeLaK cells. (A) Top: Violin plots of GFP-H2A.Z.1 (left) and GFP-PWWP2A (right) ChIP-seq read counts at HMG20A-only (blue) or HMG20A+H2A.Z (yellow) binding sites. Bottom: Venn diagram displaying numbers of GFP-H2A.Z.1, -PWWP2A and -HMG20A ChIP- seq binding sites and their overlap. (B) Validation of ChIP-seq data by ChIP-qPCR at selected loci. Shown is percent input of three biological replicates of GFP, GFP-HMG20A, -HMG or -CC ChIP-qPCR of HMG20A-only sites (red: RNUB-1 and RNUE-1downstream; see red bar in Figure 3A as example), HMG20A+H2A.Z.1-positive sites (purple: EIF4H promoter and ADAMTS3 gene body; see blue bar in Figure 3A as example) and an HMG20A/H2A.Z.1-negative site (green: RPL11 gene body; see green bar in Figure 3A as example) as negative control. Data is presented as mean ± SEM of three biological replicates. (C) Enrichment plot depicting accumulation of published STARR-seq signals4 - which correspond to enhancers - at HMG20A-only (blue) or HMG20A+H2A.Z (yellow) ChIP- seq regions. (D) Violin plots of GFP-H2A.Z.1, GFP-PWWP2A, H3K4me3 (promoter), H3K4me1 (enhancer), H3K27ac (active regulatory regions) ChIP-seq read counts at HMG20A+H2A.Z (yellow) or HMG20A-only (blue) binding sites. (E) Top-enriched motifs within HMG20A-only (top) or within HMG20A+H2A.Z (bottom) ChIP-seq peaks identified with MEME-ChIP. (F) Average binding plots of both replicates of ChIP-seq identified GFP-HMG20A binding sites. The line colors reflect the average binned expression levels of associated genes. Source data for these figures are provided as a Source Data file. | |
Supplementary Figure 4: hmg20a expression in X. laevis. (A) Alignment of human (H. sapiens), mouse (M. musculus) and frog (X. laevis) Hmg20a protein sequences. HMG box is shown in blue, coiled-coil region in red. Alignment was performed using Clustal Omega. (B) Temporal expression pattern of Xenopus hmg20a: RT-qPCR of hmg20a mRNA expression covering X. laevis developmental stages 4 (8-cell stage) to 42 normalized to odc expression. Error bars indicate s.e.m. of three technical replicates. (C-R’) Spatial expression pattern of hmg20A determined by whole mount in situ hybridization. hmg20A mRNA is detected at early stages of Xenopus laevis development. (C) 8-cell stage embryo, anterior view. (D) 8-cell stage embryo, dorso- lateral view, animal and vegetal pole are indicated. (E) Embryo at blastula stage 6.5. anterior view. (F) Same embryo as in E, dorsal view. (G) Embryo at gastrula stage 10. (H) Embryo at neurula stage 19, anterior view. (I) Same embryo as in H, dorsal view. (J) Embryo at stage 20, lateral view. (K) Embryo at stage 24, lateral view. (L) Sense control, embryo at stage 24. (M) Embryo at stage 33, lateral view. (N) Sense control, embryo at stage 33. Scale bar in C-N is 1mm. (O) Transverse section through the branchial arch region of a stage 31 embryo, hmg20A expression in the branchial arches is indicated by arrows. (P-R’) Transverse sections of a stage 42 embryo. (P) hmg20A is partially expressed in the heart region. (Q) hmg20A expression within the notochord (no). (R, R’) hmg20A is partially expressed in the brain and eye. Scale bar in O-R’ is 100 μm. abbreviations: a, atrium, an, animal; b, brain; ba, branchial arches; bl, blastoporus; ea, eye anlage; e, eye; inl, inner nuclear layer; le, lens; mn, migratory neural crest; nf, neural fold; no, notochord; ov, otic vesicle; rpe, retinal pigment epithelium v, ventricle; ve, vegetal. Source data for these figures are provided as a Source Data file. | |
hmg20a (high mobility group 20A ) gene expression in X. laevis embryo, NF stage 4 (8-cell), assayed via in situ hybridization, animal view (C) and lateral view (D) with animal pole up. | |
hmg20a (high mobility group 20A ) gene expression in X. laevis embryo, NF stage 6.5, assayed via in situ hybridization, animal view (E) and lateral view (F) with animal pole up. | |
hmg20a (high mobility group 20A ) gene expression in X. laevis embryo, NF stage 10, assayed via in situ hybridization, blasts-oral/vegetal view, dorsal up. | |
hmg20a (high mobility group 20A ) gene expression in X. laevis embryo, NF stage 19, assayed via in situ hybridization, anterior view (H) with dorsal surface up, and dorsal view (I) with anterior up. | |
hmg20a (high mobility group 20A ) gene expression in X. laevis embryo, NF stage 20, assayed via in situ hybridization, lateral view, anterior right, dorsal up. | |
hmg20a (high mobility group 20A ) gene expression in X. laevis embryo, NF stage 24, assayed via in situ hybridization, lateral view, anterior right, dorsal up | |
hmg20a (high mobility group 20A ) gene expression in transverse sections of a X. laevis embryo, NF stage 31, assayed via in situ hybridization: O. through pharyngeal/branchial arches; P. through head and eyes; Q. through mid-truck region; in all images dorsal is up. | |
hmg20a (high mobility group 20A ) gene expression in transverse and coronal sectional planes of a X. laevis embryo, NF stage 42, assayed via in situ hybridization R. through head and eye; R'. enlargement of eye. | |
Supplementary Figure 5: Loss of HMG20A impairs mESC differentiation (A) Schematic depiction of Hmg20A DP generation in mESCs by introducing mCherry_triple terminator sites and a puromycin resistance_triple terminator site into both Hmg20a alleles directly after the start codon by a CRISPR/Cas9-based approach. Created with BioRender.com. (B) RT-qPCR of EMT marker genes Slug, Twist Cdh2, pluripotency marker Oct4 and Hmg20a in WT cells at Day0 and Day9 of neural crest differentiation protocol. Expression was normalized to Hprt, 18S RNA and Gapdh expression. Data is presented as mean ± SEM of three technical replicates. (C) Migration assay. Top: Representative microscopy pictures of EBs at Day9 of neural crest differentiation protocol depicting migrating (left, see arrows) or not migrating (right) cells. Bottom: Quantification of migration capability of cells from WT and three Hmg20a DP embryoid body (EB) clones based on visual inspection (see top pictures). (Chi-square, two-sided p= 0.0344 (#06), p = 0.0301 (#26), p = 0.4260 (#48)). (D) RT-qPCR of cardiomyocyte marker genes Acta2 (middle) and Gata4 (right) in WT cells at Day2 and Day7.5 of neural crest differentiation protocol. Expression was normalized to Hprt, 18S RNA and Gapdh expression. Data is presented as mean ± SEM of three technical replicates. (E) Phase-contrast microscopy images of WT and Hmg20a DP clone #26 mESCs and EBs during cardiomyocyte differentiation. (F) Size of EBs of WT and three individual Hmg20a DP cells at Day6 of cardiomyocyte differentiation protocol. Number of measured EBs indicated above. (Man-Whitney, two-tailed p= 3.6101185369e-015 (#06), p = 5.4103237427e-023 (#26), p = 4.2379561787e-010 (#48)). Error bars indicate min/max values, lines indicate median, boxes indicate 25th to 75th percentiles. Data was derived from two independent differentiation approaches. Experiments in E, F were repeated independently two times with consistency. Source data for these figures are provided as a Source Data file. | |
Supplementary Figure 6: HMG20A regulates early transcription programs of cardiomyocyte differentiation. GO term analysis of deregulated genes upon HMG20A depletion within 10 clusters identified in Figure 6C. | |
Supplementary Figure 7: HMG20A is associated with transcriptionally active genes. (A) Density heatmap of 2,545 HMG20A binding sites detected in CUT&RUN (see Figure 7A). Color intensity represents normalized and globally scaled tag counts. (B) Average binding plots of both replicates of CUT&RUN identified HMG20A binding sites (see Supplemental Figure 7A). The line colors reflect the average binned expression levels of associated genes. (C) Gene set enrichment plot of genes associated with differentially accessible regions (DARs) after HMG20A depletion correlated to gene expression. Notice that more open accessibly sites correlate with increase in gene transcription (left; padj = 5.502329e- 16; NES = 2.2991133), while more inaccessible sites correlate with reduction in gene expression (right; padj = 1.620638e-04; NES = -1.6649125)5. | |
Supplementary Figure 8: HMG20A localizes to specific DNA-sequences in mESCs Top-enriched motifs within HMG20A-only (top) or within HMG20A+H2A.Z (bottom) CUT&RUN peaks identified with MEME-ChIP. |
References [+] :
Achilleos,
Neural crest stem cells: discovery, properties and potential for therapy.
2012, Pubmed
Achilleos, Neural crest stem cells: discovery, properties and potential for therapy. 2012, Pubmed
Akerberg, A reference map of murine cardiac transcription factor chromatin occupancy identifies dynamic and conserved enhancers. 2019, Pubmed
Amemiya, The ENCODE Blacklist: Identification of Problematic Regions of the Genome. 2019, Pubmed
Bailey, Fitting a mixture model by expectation maximization to discover motifs in biopolymers. 1994, Pubmed
Barriga, In Vivo and In Vitro Quantitative Analysis of Neural Crest Cell Migration. 2019, Pubmed , Xenbase
Bornelöv, The Nucleosome Remodeling and Deacetylation Complex Modulates Chromatin Structure at Sites of Active Transcription to Fine-Tune Gene Expression. 2018, Pubmed
Bönisch, Chromatin proteomics and epigenetic regulatory circuits. 2008, Pubmed
Bönisch, H2A.Z.2.2 is an alternatively spliced histone H2A.Z variant that causes severe nucleosome destabilization. 2012, Pubmed
Ceballos-Chávez, Control of neuronal differentiation by sumoylation of BRAF35, a subunit of the LSD1-CoREST histone demethylase complex. 2012, Pubmed
Colino-Sanguino, The H2A.Z-nuclesome code in mammals: emerging functions. 2022, Pubmed
Cox, Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. 2014, Pubmed
Eberl, A map of general and specialized chromatin readers in mouse tissues generated by label-free interaction proteomics. 2013, Pubmed
Edgar, Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. 2002, Pubmed
Eirín-López, The evolutionary differentiation of two histone H2A.Z variants in chordates (H2A.Z-1 and H2A.Z-2) is mediated by a stepwise mutation process that affects three amino acid residues. 2009, Pubmed
Ernst, ChromHMM: automating chromatin-state discovery and characterization. 2012, Pubmed
Ernst, Chromatin-state discovery and genome annotation with ChromHMM. 2017, Pubmed
Faast, Histone variant H2A.Z is required for early mammalian development. 2001, Pubmed
Garay, Yin-yang actions of histone methylation regulatory complexes in the brain. 2016, Pubmed
Gentleman, Bioconductor: open software development for computational biology and bioinformatics. 2004, Pubmed
Giaimo, The histone variant H2A.Z in gene regulation. 2019, Pubmed
Gómez-Marín, The high mobility group protein HMG20A cooperates with the histone reader PHF14 to modulate TGFβ and Hippo pathways. 2022, Pubmed
H Backman, systemPipeR: NGS workflow and report generation environment. 2016, Pubmed
Hahne, Visualizing Genomic Data Using Gviz and Bioconductor. 2016, Pubmed
Hirota, The nucleosome remodeling and deacetylase complex protein CHD4 regulates neural differentiation of mouse embryonic stem cells by down-regulating p53. 2019, Pubmed
Iouzalen, H2A.ZI, a new variant histone expressed during Xenopus early development exhibits several distinct features from the core histone H2A. 1996, Pubmed , Xenbase
Kawaguchi, Osteogenic and chondrogenic differentiation of embryonic stem cells in response to specific growth factors. 2005, Pubmed
Kim, HISAT: a fast spliced aligner with low memory requirements. 2015, Pubmed
Kreienbaum, H2A.Z's 'social' network: functional partners of an enigmatic histone variant. 2022, Pubmed
Laugesen, Chromatin repressive complexes in stem cells, development, and cancer. 2014, Pubmed
Lawrence, Software for computing and annotating genomic ranges. 2013, Pubmed
Link, PWWP2A binds distinct chromatin moieties and interacts with an MTA1-specific core NuRD complex. 2018, Pubmed
Link, PWWP2A: A novel mitosis link? 2017, Pubmed
Litviňuková, Cells of the adult human heart. 2020, Pubmed
Love, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. 2014, Pubmed
Low, The Nucleosome Remodeling and Deacetylase Complex Has an Asymmetric, Dynamic, and Modular Architecture. 2020, Pubmed
Lynch, Cardiomyocyte Differentiation from Mouse Embryonic Stem Cells. 2018, Pubmed
McClellan, Growth Factor Independence 1B-Mediated Transcriptional Repression and Lineage Allocation Require Lysine-Specific Demethylase 1-Dependent Recruitment of the BHC Complex. 2019, Pubmed
Megason, Digitizing life at the level of the cell: high-performance laser-scanning microscopy and image analysis for in toto imaging of development. 2003, Pubmed
Meneghini, Conserved histone variant H2A.Z protects euchromatin from the ectopic spread of silent heterochromatin. 2003, Pubmed
Moriyoshi, Labeling neural cells using adenoviral gene transfer of membrane-targeted GFP. 1996, Pubmed
Muerdter, Resolving systematic errors in widely used enhancer activity assays in human cells. 2018, Pubmed
Mulas, Defined conditions for propagation and manipulation of mouse embryonic stem cells. 2019, Pubmed
Nicholson, A hypomorphic lsd1 allele results in heart development defects in mice. 2013, Pubmed
Perez-Riverol, The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. 2022, Pubmed
Procida, JAZF1, A Novel p400/TIP60/NuA4 Complex Member, Regulates H2A.Z Acetylation at Regulatory Regions. 2021, Pubmed
Pünzeler, Multivalent binding of PWWP2A to H2A.Z regulates mitosis and neural crest differentiation. 2017, Pubmed , Xenbase
Qureshi, REST and CoREST are transcriptional and epigenetic regulators of seminal neural fate decisions. 2010, Pubmed
Ramírez, deepTools: a flexible platform for exploring deep-sequencing data. 2014, Pubmed
Rangasamy, RNA interference demonstrates a novel role for H2A.Z in chromosome segregation. 2004, Pubmed
Rivera, Unveiling RCOR1 as a rheostat at transcriptionally permissive chromatin. 2022, Pubmed
Rivero, HMG20A is required for SNAI1-mediated epithelial to mesenchymal transition. 2015, Pubmed
Santisteban, Histone H2A.Z regulats transcription and is partially redundant with nucleosome remodeling complexes. 2000, Pubmed
Schwenty-Lara, The histone methyltransferase KMT2D, mutated in Kabuki syndrome patients, is required for neural crest cell formation and migration. 2020, Pubmed , Xenbase
Schwenty-Lara, Loss of function of Kmt2d, a gene mutated in Kabuki syndrome, affects heart development in Xenopus laevis. 2019, Pubmed , Xenbase
Silva, The N-terminal Region of Chromodomain Helicase DNA-binding Protein 4 (CHD4) Is Essential for Activity and Contains a High Mobility Group (HMG) Box-like-domain That Can Bind Poly(ADP-ribose). 2016, Pubmed
Stros, The HMG-box: a versatile protein domain occurring in a wide variety of DNA-binding proteins. 2007, Pubmed
Torrado, Refinement of the subunit interaction network within the nucleosome remodelling and deacetylase (NuRD) complex. 2017, Pubmed
Tripathi, Mechanistic regulation of epithelial-to-mesenchymal transition through RAS signaling pathway and therapeutic implications in human cancer. 2018, Pubmed
Tyanova, The Perseus computational platform for comprehensive analysis of (prote)omics data. 2016, Pubmed
Ufartes, De novo mutations in FBRSL1 cause a novel recognizable malformation and intellectual disability syndrome. 2020, Pubmed , Xenbase
Ungefroren, The role of small GTPases of the Rho/Rac family in TGF-β-induced EMT and cell motility in cancer. 2018, Pubmed
Vardabasso, Histone Variant H2A.Z.2 Mediates Proliferation and Drug Sensitivity of Malignant Melanoma. 2015, Pubmed
Wei, The PWWP2A Histone Deacetylase Complex Represses Intragenic Spurious Transcription Initiation in mESCs. 2020, Pubmed
Welcker, Insm1 controls development of pituitary endocrine cells and requires a SNAG domain for function and for recruitment of histone-modifying factors. 2013, Pubmed
Wilczewski, CHD4 and the NuRD complex directly control cardiac sarcomere formation. 2018, Pubmed
Wratting, A conserved function for the H2A.Z C terminus. 2012, Pubmed
Wynder, Recruitment of MLL by HMG-domain protein iBRAF promotes neural differentiation. 2005, Pubmed
Yamamoto, Identification and Biochemical Characterization of High Mobility Group Protein 20A as a Novel Ca2+/S100A6 Target. 2021, Pubmed
Yu, ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. 2015, Pubmed
Zhang, A variant NuRD complex containing PWWP2A/B excludes MBD2/3 to regulate transcription at active genes. 2018, Pubmed
Zhang, Model-based analysis of ChIP-Seq (MACS). 2008, Pubmed
Zhou, Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. 2019, Pubmed
Zovkic, Histone H2A.Z subunit exchange controls consolidation of recent and remote memory. 2014, Pubmed
van Beest, Sequence-specific high mobility group box factors recognize 10-12-base pair minor groove motifs. 2000, Pubmed
van Daal, A histone variant, H2AvD, is essential in Drosophila melanogaster. 1992, Pubmed
van de Nobelen, CTCF regulates the local epigenetic state of ribosomal DNA repeats. 2010, Pubmed