Zachary D. Smith’s research while affiliated with Yale-New Haven Hospital and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (74)


Crossfirre, Firre, and Dxz4 loci are the topmost female-specific loci by chromatin accessibility with unique allele-specific characteristics
a Genome browser tracks of the mouse brain showing strand-specific RNA-seq¹⁵ and ATAC-seq²³ for females (red) and males (blue) covering the Crossfirre (Gm35612), Firre, and Dxz4 (4933407K13Rik) locus. The gene body of Crossfirre is embedded in a 50 Kb LINE cluster. Boxes highlight female-specific ATAC-seq peaks identified through MACS2⁶⁰. ENCODE Candidate Cis-Regulator Elements (cCRES) are shown for each locus (Promoter: red, Proximal enhancer: orange, Distal enhancer: yellow, DNase/H3K4me3: pink, CTCF: blue). b Overview of six female and male organs (two biological replicates per sex, n = 24) utilized to map sex-specific chromatin accessible loci from publicly available ATAC-seq data²³ (left). A one-sided binomial test was used to identify sex-specific loci genome-wide by comparing male and female ATAC peak counts binned over 100 Kb sliding windows (right, see section “Methods”). A positive value was assigned if more peaks were present in females than in males, while negative values were assigned if the reverse was true. The top female-specific loci Crossfirre, Firre, and Dxz4 are indicated, together with the expected female-specific Xist locus. c Sex-specific analysis of (b) for the entire X chromosome. d Violin plots displaying the allelic ratio of ATAC enrichment over a 50 Kb sliding window for the entire X chromosome of neuronal progenitor cell clones (n = 2)²⁴. The red dot highlights the allelic ratio of the sliding window overlapping the Crossfirre/Firre and Dxz4 locus. Boxes represent the interquartile range around the median, while the whiskers extend to 1.5 times the interquartile range. e RNA-sequencing abundance of Crossfirre (red), Firre (orange), and Dxz4 (gray) across various mouse organs. Shown are the log10-transformed mean TPM values of each gene within the respective tissue with a pseudocount of 1 added.
Mice carrying a Crossfirre single deletion or combined with Firre and Dxz4 are viable and undergo normal development
a Schematic overview of the X chromosome. The megadomains and the superloop between the Firre and Dxz4 loci are specific to the inactive X chromosome (Xi), whereas full-length transcription of Firre and Dxz4 only occurs on the active X chromosome (Xa). The imprinted lncRNA Crossfirre shows expression from the maternal X chromosome, independent of random XCI¹⁵. Dotted lines indicate the deleted loci for ∆Firre (orange), ∆Dxz4 (gray), ∆Firre-Dxz4 (blue), ∆Crossfirre (red), ∆Crossfirre-Firre (green), ∆Crossfirre-Firre-Dxz4 (TKO, turquoise). Colors are used in figures throughout the manuscript to highlight the genotype of origin. White stars refer to previously generated mouse strains¹⁴. b Genome browser RNA-seq tracks showing the Crossfirre/Firre (left) and Dxz4 (right) locus for the adult spleen of wildtype (black), and ∆Crossfirre, ∆Crossfirre-Firre, TKO mutants. Scissors indicate the start and end of the CRISPR-Cas9 deletion (see section “Methods”). c Mean expression values of Crossfirre, Firre, and Dxz4 in the adult spleens of Crossfirre mutant strains (WT n = 4, ∆Crossfirre n = 3, ∆Crossfirre-Firre n = 2, TKO n = 3). Error bars indicate the standard deviation. d Sex distribution of homozygous ∆Crossfirre, ∆Crossfirre-Firre, and TKO breeding. The p values are obtained from a one-sided binomial test.
Deleting the imprinted Crossfirre locus alone or together with Firre and Dxz4 does not affect imprinted XCI
a Schematic of our experimental system to investigate the impact of the deletions on the inactive X (Xi, left) or active X (Xa, right) for imprinted X inactivation. RNA-seq data of E12.5 female placentas are analyzed from wildtype (WT) F1 reciprocal crosses (CASTxBL6 n = 9, BL6xCAST n = 8), and for the six F1 mutants carrying the deletion on the paternal Xi (n = 3 per genotype) or on the maternal Xa (n = 3 per genotype). The relative expression (mean and standard deviation) between WT and ∆Crossfirre-Firre-Dxz4 (TKO, turquoise) is shown for deletions on Xi (left) and Xa (right). b Number of differentially expressed genes across mutant strains (DEseq2: FDR ≤ 0.01, |log2FC| ≥ 1). c FDR-adjusted P values of differential gene expression analysis between WT and TKO on Xi (left) and Xa (right). Venn diagram showing the overlap of dysregulated genes between ∆Firre-Dxz4 (blue), ∆Crossfirre-Firre (green), and the TKO (turquoise). d Median allelic ratios for X-linked genes in WT (black) and the six knockout strains carrying the deletions on Xi (left) or Xa (right). The blue dot emphasizes the paternal allelic ratio of the lncRNA Xist. The allelic ratios range from 0 to 1 such that 1 corresponds to maternal expression (MAT), 0.5 to biallelic expression, and 0 to paternal expression (PAT). Boxes indicate the interquartile range around the median and whiskers 1.5x the interquartile range. e Heatmap showing median allelic ratios for X-linked genes that are informative across all WT and knockout strains carrying the deletions on Xi (upper panel) or Xa (lower panel). Brown indicates an allelic ratio of 1 (CAST allele), while black indicates an allelic ratio of 0 (BL6 allele). Common escape genes Kdm6a and Eif2s3x are highlighted showing biallelic expression, thus validating our approach. Arrows indicate the approximate location of Crossfirre, Firre, and Dxz4. *The expression of Tsix from Xi is due to the overlapping nature with Xist and thus an artifact of the non-stranded analysis.
Deletion of topmost female-specific loci Crossfirre, Firre, and Dxz4 does not affect random XCI
a Schematic overview of the experimental setup to investigate the effects of the ∆Crossfirre-Firre-Dxz4 (TKO) on the active X (Xa) or inactive X (Xi) upon random XCI. Heterozygous TKO females (BL6) were mated with wildtype (WT) CAST mice to generate F1 hybrids with WT and heterozygous TKO genotypes. Spleens were isolated (WT n = 1, TKO heterozygous n = 1) and processed for single-cell RNA-seq. Sequencing data were subjected to bioinformatic preprocessing, including alignment, quality control, and normalization. The dataset was split according to the genotype condition (WT/TKO). b UMAP of unsupervised clustering by genotype condition (WT/TKO). c Single-cell RNA-seq data were further split by Xa chromosome state (CAST Xa, BL6 Xa) using Allelome.PRO and a chromosome-wide window (see section “Methods”). Thus, we obtained single-cell RNA-seq data from WT and TKO spleen cells which could be categorized in one of four groups: WT CAST Xa, WT BL6 Xa, TKO Xi (CAST Xa), and TKO Xa (BL6 Xa). d Heatmap showing the median allelic ratios for informative X-linked genes in WT and TKO mice carrying the deletions on Xi (left) or Xa (right). Read counts of all cells were summarized as pseudobulk to increase gene coverage. The brown color indicates an allelic ratio of 1 corresponding to the CAST allele, while black indicates an allelic ratio of 0 (BL6 allele). Arrows indicate the approximate location of Crossfirre, Firre, and Dxz4. *The expression of Tsix from Xi is due to the overlapping nature with Xist and thus an artifact of the non-stranded analysis.
Homozygous double deletion of Crossfirre-Firre, results in upregulation of mitochondrial and ribosomal pathways
a Transcriptomic bodymap for six different organs from homozygous adult female ∆Firre-Dxz4 and ∆Crossfirre-Firre-Dxz4 (TKO) mice (wildtype n = 4; ∆Firre-Dxz4 n = 4; TKO n = 3). Chart bars show the number of significantly differentially expressed genes in the spleen, kidney, lung, heart, liver, and brain for TKO and ∆Firre-Dxz4 (DEseq2: FDR ≤ 0.01, |log2FC| ≥ 1). Pie plots represent the proportion of differentially expressed genes between the X chromosome and autosomes as percentages. The size of the pie plots is proportional to the total amount of differentially expressed genes. b Number of differentially expressed genes shared across two, three, four, five, and six different tissues in TKO mice. The heatmap below shows the log2fold changes of the overlapping dysregulated genes per tissue being up- (orange) or downregulated (black). c Heatmap showing the log10(FDR) of the top 100 significantly enriched gene sets from TKO gene set enrichment analysis (left; FDR ≤ 0.1). Network plot for spleen showing 18 gene set clusters with two dominant groups associated with mitochondrial (cluster ID: 1 n = 21) and ribosomal (cluster ID: 3 n = 35) gene sets (right). d Differential gene expression results from RNA-seq data obtained from female spleens of the different knockout strains. The number of significantly up- and downregulated genes is shown per genotype. e Number of significantly differentially expressed genes unique for each genotype and shared by the different knockout models. Below, heatmap showing log2fold changes of differentially expressed genes shared between ∆Crossfirre-Firre and TKO mice. Color shading indicates up- (orange) and downregulation (black). f Gene set enrichment analysis of dysregulated genes from TKO, ∆Crossfirre-Firre, ∆Dxz4, ∆Firre, and ∆Crossfirre deletions. The heatmap shows the log10(FDR) of all informative gene sets of the top 100 significantly enriched gene sets detected in (c).

+1

X-linked deletion of Crossfirre, Firre, and Dxz4 in vivo uncovers diverse phenotypes and combinatorial effects on autosomes
  • Article
  • Full-text available

December 2024

·

58 Reads

Tim P. Hasenbein

·

·

Zachary D. Smith

·

[...]

·

The lncRNA Crossfirre was identified as an imprinted X-linked gene, and is transcribed antisense to the trans-acting lncRNA Firre. The Firre locus forms an inactive-X-specific interaction with Dxz4, both loci providing the platform for the largest conserved chromatin structures. Here, we characterize the epigenetic profile of these loci, revealing them as the most female-specific accessible regions genome-wide. To address their in vivo role, we perform one of the largest X-linked knockout studies by deleting Crossfirre, Firre, and Dxz4 individually and in combination. Despite their distinct epigenetic features observed on the X chromosome, our allele-specific analysis uncovers these loci as dispensable for imprinted and random X chromosome inactivation. However, we provide evidence that Crossfirre affects autosomal gene regulation but only in combination with Firre. To shed light on the functional role of these sex-specific loci, we perform an extensive standardized phenotyping pipeline and uncover diverse knockout and sex-specific phenotypes. Collectively, our study provides the foundation for exploring the intricate interplay of conserved X-linked loci in vivo.

Download

DNA methylation in mammalian development and disease

August 2024

·

35 Reads

·

17 Citations

Nature Reviews Genetics

The DNA methylation field has matured from a phase of discovery and genomic characterization to one seeking deeper functional understanding of how this modification contributes to development, ageing and disease. In particular, the past decade has seen many exciting mechanistic discoveries that have substantially expanded our appreciation for how this generic, evolutionarily ancient modification can be incorporated into robust epigenetic codes. Here, we summarize the current understanding of the distinct DNA methylation landscapes that emerge over the mammalian lifespan and discuss how they interact with other regulatory layers to support diverse genomic functions. We then review the rising interest in alternative patterns found during senescence and the somatic transition to cancer. Alongside advancements in single-cell and long-read sequencing technologies, the collective insights made across these fields offer new opportunities to connect the biochemical and genetic features of DNA methylation to cell physiology, developmental potential and phenotype.


A second wave of Notch signaling diversifies the intestinal secretory lineage

July 2024

·

10 Reads

The small intestine is well known for the function of its nutrient-absorbing enterocytes; yet equally critical for the maintenance of homeostasis is a diverse set of secretory cells, all of which are presumed to differentiate from the same intestinal stem cell. Despite major roles in intestinal function and health, understanding how the full spectrum of secretory cell types arises remains a longstanding challenge, largely due to their comparative rarity. Here, we investigate the fate specification of a rare and distinct population of small intestinal epithelial cells found in rats and humans but not mice: CFTR High Expressers (CHEs). We use pseudotime trajectory analysis of single-cell RNA-seq data from rat intestinal jejunum to provide evidence that CHEs are specified along the secretory lineage and appear to employ a second wave of Notch-based signal transduction to distinguish these cells from other secretory cell types. We further validate the general order of transcription factors that direct these cells from unspecified progenitors within the crypt and experimentally demonstrate that Notch signaling is necessary to induce CHE fate both in vivo and in vitro. Our results suggest a model in which Notch is reactivated along the secretory lineage to specify the CHE population: a rare secretory cell type with putative functions in localized coordination of luminal pH and direct relevance to cystic fibrosis pathophysiology.



Autonomous transposons tune their sequences to ensure somatic suppression

February 2024

·

129 Reads

·

10 Citations

Nature

Transposable elements (TEs) are a major constituent of human genes, occupying approximately half of the intronic space. During pre-messenger RNA synthesis, intronic TEs are transcribed along with their host genes but rarely contribute to the final mRNA product because they are spliced out together with the intron and rapidly degraded. Paradoxically, TEs are an abundant source of RNA-processing signals through which they can create new introns¹, and also functional² or non-functional chimeric transcripts³. The rarity of these events implies the existence of a resilient splicing code that is able to suppress TE exonization without compromising host pre-mRNA processing. Here we show that SAFB proteins protect genome integrity by preventing retrotransposition of L1 elements while maintaining splicing integrity, via prevention of the exonization of previously integrated TEs. This unique dual role is possible because of L1’s conserved adenosine-rich coding sequences that are bound by SAFB proteins. The suppressive activity of SAFB extends to tissue-specific, giant protein-coding cassette exons, nested genes and Tigger DNA transposons. Moreover, SAFB also suppresses LTR/ERV elements in species in which they are still active, such as mice and flies. A significant subset of splicing events suppressed by SAFB in somatic cells are activated in the testis, coinciding with low SAFB expression in postmeiotic spermatids. Reminiscent of the division of labour between innate and adaptive immune systems that fight external pathogens, our results uncover SAFB proteins as an RNA-based, pattern-guided, non-adaptive defence system against TEs in the soma, complementing the RNA-based, adaptive Piwi-interacting RNA pathway of the germline.


HEEs reproducibly model post-implantation-like lineage bifurcation
a, Schematic of hEE generation. SDM, spontaneous differentiation medium. SDM, spontaneous differentiation media. Y2, Y27632. mIVC2, modified in vitro culture 2 media. b, Time-course development of hEEs. Scale bar, 50 µm. n = 11, 6 and 7 independent experiments from RUES2, H9 and ESI017 hPS cells, respectively. c, Schematic of human embryo development at the indicated Carnegie stages (CS). d, Sampled frames from a timelapse movie of hEE organization (top). n = 30 structures; n = 3 independent experiments. Scale bar, 20 µm. Phase-contrast image of D5 hEEs (bottom). The inset image highlights the inner cavity (red circle), epiblast-like (blue circle) and hypoblast-like (white circle) compartments. n ≥ 30 independent experiments. Scale bar, 20 µm. e, Percentage of SOX2⁺ and SOX17⁺ cells in hEEs. n = 20 structures per timepoint. Each dot represents one structure. The plot shows the median (thick solid line) and quartiles (thin dotted line). f, hEE efficiency versus aggregates comprising a single compartment (embryonic-like (E) and extra-embryonic-like (Ex.E)). Plots show mean ± s.d. n = 1,764 structures from D4 and D5. n = 24 independent experiments for hEE and 13 independent experiments for embryonic-like only and extra-embryonic-like only (for the noted genetic background variation, see Supplementary Table 2). g, Histological section of a CS5b stage in vivo human embryo (left; obtained from the Virtual Human Embryo project) compared with an in vitro hEE at D5 (right). Scale bar, 50 µm. n = 9 independent experiments. h, Same structure as panel g but with inverted and enhanced N-cadherin (NCAD) intensity for better clarity (left). An ozone graph of nuclear length and height of presumptive amnion-like (red) and epiblast-like (blue) cells is also shown (right). i, Chimeric integration of hEE-derived SOX17–tdTomato cells into primitive (top) or visceral endoderm (bottom) of mouse E4.5 blastocysts or E5.5 embryos. n = 15/54 blastocysts and 4/10 E5.5 embryos. n = 2 independent experiments. Scale bars, 20 µm. The arrows indicate the successful integration of human SOX17-TdTomato cells into the mouse primitive or visceral endoderm. j, Expansion of SOX17–tdTomato cells in 2D culture, which were sorted from D3 or D4 hEEs. The red lines outline cell colonies, which are also shown in the zoomed-in images. n = 3 independent experiments. Scale bars, 200 µm. Illustrations in a,c,i, credit: A.L. Cox. The embryo section in g, courtesy of the Virtual Human Embryo.
Source Data
The emergence of perigastrulation lineages in hEEs
a, 3D uniform manifold approximation and projection (UMAP) plot (top), pie charts (bottom) and gene expression heatmap (right) of D4 and D6 structures. n = 18,042 total cells. G1 Hypo, growth 1 hypoblast-like; G2M/S Hypo, growth 2 mitosis/synthesis hypoblast-like; meso-like, mesoderm-like; PS-like, primitive streak-like. b, Dot plot of mean marker gene expression level. c, Integrated reference of three studies of primate development (left). See Methods and Supplementary Notes. Projection of hEE scRNA-seq data (query) onto in vivo reference is also shown (right). hEE cells are annotated as above. Reference cells are in grey to represent the background distribution of primate states. For the in vivo reference lineage abbreviations, refer to Extended Data Fig. 3d and Supplementary Table 3. d, hEEs generated from SMAD1–RFP hPS cells (top). Scale bar, 20 µm. The inner compartment (dotted lines, schematics) ISL1 phenotype frequency is also shown (bottom). Each dot represents the percentage of structures per tile scan. The plot shows mean ± s.d. n = 346 structures from 2 independent experiments specific to the RUES2 background are presented in the graph. n = 13 experiments total across different genetic backgrounds. e, SMAD1–RFP 3D surface intensity plot (same structure as shown in panel d) (left). Scale bar, 20 µm. A scatter plot of nuclear SMAD1–RFP fluorescence intensity in ISL1⁻ and ISL1⁺ cells. n = 1,677 cells in 3 representative structures from 2 independent experiments. Two-tailed unpaired, parametric t-test with Welsch’s correction. P values are displayed in the figure. f, Percentage of ISL1⁺ structures (RUES2 background). D4 and D5 control (n = 489), BMP2 (n = 461), BMP4 (n = 405), BMP7 (n = 391) and LDN (n = 521). Three or four independent experiments per group. Post-hoc Dunnett’s multiple comparison test, one-way ANOVA. P values are displayed in the figure. Mean ± s.d. are shown.
Source Data
TGFβ and FGF signalling stabilizes hypoblast-like specification and embryo-like morphogenesis
a, GM130 (n = 169) and PODXL (n = 156) immunofluorescence within D3 hEEs. Four independent experiments each. Scale bars, 50 μm. Yellow arrowheads indicate the expression of apical markers. b, Dot plot of basement membrane gene expression in hEEs. For lineage abbreviations, refer to Fig. 2a. c, Schematic of mTeSR hPS cell aggregation strategies (top), their corresponding D4 structures (bottom left) and the cavitated hEE efficiency (bottom right). The yellow arrowheads indicate multiple cavities. Plots show mean ± s.d. n = 439 structures. Data are from four technical replicates and two independent experiments. Scale bars, 20 μm. d, UMAP plots of yolk sac endoderm genes within the D6 hypoblast-like cluster. e, AFP⁺ staining (arrowheads). The yellow and white dashed lines indicate epiblast-like and yolk sac-like compartment patterning, respectively. n = 15 structures over 2 independent experiments. Scale bar, 20 μm. f, Inhibitor treatment strategy (top; see Methods) and resulting phenotypes (bottom). The n numbers are indicated in panel g. Scale bar, 20 μm. g, Percentage of structures that specify hypoblast-like lineage. Control (C; n = 220), XAV939 (XAV; n = 101), IWP2 (IWP; n = 132; 2 experiments), SB431542 (SB; n = 125), PD0325901 (PD; n = 66) and SU5402 (SU; n = 109). The plot shows mean ± s.d. Data are from three independent experiments. Post-hoc Dunnett’s multiple comparison test, one-way ANOVA. P values are shown in the figure. Each dot represents an independent experiment. h, Cavitation efficiency per treatment. Control (n = 140), XAV (n = 95), IWP (n = 118; 2 experiments), SB (n = 79), PD (n = 66) and SU (n = 109). Data are from three independent experiments, unless otherwise indicated. The plot shows mean ± s.d. The same test as panel g was applied. P values are shown in the figure. Each dot represents an independent experiment. i, Number of SOX2⁺ or SOX17⁺ cells per structure and their percentage after each treatment. Each dot represents one structure. Control (n = 21), XAV (n = 34), IWP (n = 16), SB (n = 30), PD (n = 35) and SU (n = 30). Data are from two or more independent experiments. The plot shows the median (thick solid line) and quartiles (thin dotted line). The same test as panel g was applied. P values are shown in the figure. Illustrations in c,f, credit: A.L. Cox.
Source Data
HEEs recapitulate key hallmarks of human perigastrulation
a, 3D projections of T, OCT4 and CER1 (arrowheads) expression. The dashed lines demarcate mesodermal-like, epiblast-like and hypoblast-like regions. Note that the CER1⁺ signal in T⁺ cells is expected due to the maturing mesodermal state. n = 176 structures, 6 independent experiments. Scale bar, 20 µm. b, Angular distribution (left) of CER1⁺ cells relative to T⁺ cells (right; single section on the top and 3D projection on the bottom). Each vector represents the angle for one CER1⁺ cell. The vector length corresponds to the distance between a CER1⁺ cell and the midpoint of the bisecting line (dashed line). Scale bar, 50 µm. c, Phenotype frequencies after LDN or BMP4 treatments (left) and the corresponding representative structures (right). Control (n = 160), LDN 1 µM (n = 64), LDN 2 and 4 µM (n = 104), BMP4 100 ng ml⁻¹ (n = 67) and BMP4 200 and 400 ng ml⁻¹ (n = 90). Three or more independent experiments (per group) specific to the H9 background are presented in the graph. Six or more total experiments were conducted across different genetic backgrounds. Mean ± s.d. Post-hoc Dunnett’s multiple comparison test, one-way ANOVA. P values are shown in the figure. Each dot represents an independent experiment. Scale bar, 20 μm. d, T expression in D6 hEE. The zoomed-in images highlight T⁺, NCAD⁺ and SNAI1⁺ cells (arrowheads). The double-headed arrowheads show nuclear reorientation of T⁺ and SNAI1⁺ cells. The white and red dashed lines enclose epiblast-like and hypoblast-like regions, respectively. n = 20/53 T⁺ structures, 4 experiments. Scale bar, 20 μm. e, SNAI⁺ cells are peripheral to the inner compartment (white box), show downregulated E-cadherin (ECAD), breach the basement membrane (laminin; arrowheads) and focally migrate from the epiblast-like compartment. n = 32/55 T⁺ structures, 3 experiments. Scale bar, 20 μm. f, Principal curves of hallmark gastrulation markers over pseudotime (post-implantation epiblast-like to primitive streak-like to mesoderm-like states) in hEEs. PC, principal component. Dashed red line indicates PC, grey background indicates LOESS function (locally estimated scatterplot smoothing). g, Proposed mechanism of early human development as modelled in hEEs. Also note Fig. 2d for observed spontaneous heterogeneity in the amnion-like specification in hEEs. CTB, cytotrophoblast. Illustrations in g, credit: A.L. Cox.
Source Data
Self-patterning of human stem cells into post-implantation lineages

June 2023

·

319 Reads

·

75 Citations

Nature

Investigating human development is a significant scientific challenge due to the technical and ethical limitations of working with embryonic samples. In the face of these difficulties, stem cells have provided an alternative to experimentally model inaccessible stages of human development in vitro1-13. Here, we show that human pluripotent stem cells can be triggered to self-organise into three-dimensional structures that recapitulate some key spatiotemporal events of early human post-implantation embryonic development. Importantly, our system reproducibly captures spontaneous differentiation and co-development of embryonic epiblast and extra-embryonic hypoblast-like lineages, establishes key signalling hubs with secreted modulators, and can undergo symmetry breaking-like events. Single-cell transcriptomics confirms differentiation into diverse cell states of the peri-gastrulating human embryo14,15 without establishing placental cell types, including signatures of post-implantation epiblast, amniotic ectoderm, primitive streak, mesoderm, early extra-embryonic endoderm, as well as initial yolk sac induction. Collectively, our system captures key features of human embryonic development spanning from Carnegie-stage16 (CS) 4 to CS7, offering a reproducible, tractable, and scalable experimental platform to understand the basic cellular and molecular mechanisms that underlie human development, including new opportunities to dissect congenital pathologies with high throughput.


DOT1L bridges transcription and heterochromatin formation at mammalian pericentromeres

June 2023

·

43 Reads

·

9 Citations

EMBO Reports

Repetitive DNA elements are packaged in heterochromatin, but many require bursts of transcription to initiate and maintain long-term silencing. The mechanisms by which these heterochromatic genome features are transcribed remain largely unknown. Here, we show that DOT1L, a conserved histone methyltransferase that modifies lysine 79 of histone H3 (H3K79), has a specialized role in transcription of major satellite repeats to maintain pericentromeric heterochromatin and genome stability. We find that H3K79me3 is selectively enriched relative to H3K79me2 at repetitive elements in mouse embryonic stem cells (mESCs), that DOT1L loss compromises pericentromeric satellite transcription, and that this activity involves possible coordination between DOT1L and the chromatin remodeler SMARCA5. Stimulation of transcript production from pericentromeric repeats by DOT1L participates in stabilization of heterochromatin structures in mESCs and cleavage-stage embryos and is required for preimplantation viability. Our findings uncover an important role for DOT1L as a bridge between transcriptional activation of repeat elements and heterochromatin stability, advancing our understanding of how genome integrity is maintained and how chromatin state is set up during early development.


DOT1L promotes spermatid differentiation by regulating expression of genes required for histone-to-protamine replacement

April 2023

·

59 Reads

·

3 Citations

Development

Unique chromatin remodeling factors orchestrate dramatic changes in nuclear morphology during differentiation of the mature sperm head. A critical step in this process is histone-to-protamine exchange, which must be executed correctly to avoid sperm DNA damage, embryonic lethality, and male sterility. Here, we define an essential role for the histone methyltransferase DOT1L in the histone-to-protamine transition. We show that DOT1L is abundantly expressed in meiotic and postmeiotic germ cells and that methylation of histone H3 lysine 79 (H3K79), the modification catalyzed by DOT1L, is enriched in developing spermatids in the initial stages of histone replacement. Elongating spermatids lacking DOT1L fail to fully replace histones and exhibit aberrant protamine recruitment, resulting in deformed sperm heads and male sterility. Loss of DOT1L results in transcriptional dysregulation coinciding with the onset of histone replacement and affecting genes required for histone-to-protamine exchange. DOT1L also deposits H3K79me2 and promotes accumulation of elongating RNA Polymerase II at the testis-specific bromodomain gene Brdt. Together, our results indicate that DOT1L is an important mediator of transcription during spermatid differentiation and an indispensable regulator of male fertility.


Dynamic antagonism between key repressive pathways maintains the placental epigenome

April 2023

·

230 Reads

·

11 Citations

Nature Cell Biology

DNA and Histone 3 Lysine 27 methylation typically function as repressive modifications and operate within distinct genomic compartments. In mammals, the majority of the genome is kept in a DNA methylated state, whereas the Polycomb repressive complexes regulate the unmethylated CpG-rich promoters of developmental genes. In contrast to this general framework, the extra-embryonic lineages display non-canonical, globally intermediate DNA methylation levels, including disruption of local Polycomb domains. Here, to better understand this unusual landscape’s molecular properties, we genetically and chemically perturbed major epigenetic pathways in mouse trophoblast stem cells. We find that the extra-embryonic epigenome reflects ongoing and dynamic de novo methyltransferase recruitment, which is continuously antagonized by Polycomb to maintain intermediate, locally disordered methylation. Despite its disorganized molecular appearance, our data point to a highly controlled equilibrium between counteracting repressors within extra-embryonic cells, one that can seemingly persist indefinitely without bistable features typically seen for embryonic forms of epigenetic regulation.


TRIM28 degradation leads to the reduction of SE transcription and loss of transcriptional condensates at SEs in mESCs
a, Models of the TRIM28/HP1α pathway and enhancers. b, Heatmap of ChIP–seq read densities within a 2-kb window around full-length IAP ERVs and enhancers in mESC. The genomic elements were length normalized. Enhancers include the constituent enhancers of SEs and typical enhancers. Rpm, reads per million. c, Scheme of the dTAG system to degrade TRIM28 in mESCs. d, Western blot validation of the FKBP degron tag and its ability to degrade TRIM28. e, FC in read density of TT-SLAM-seq and RNA-seq data after the indicated duration of dTAG-13 treatment, normalized to the level in the DMSO control. Data are presented as mean values ± s.d. from three biological replicates. P values are from unpaired two-sided t-tests. **P < 0.01. f, Genome browser tracks of ChIP–seq data (H3K27Ac, OCT4, SOX2, NANOG) in control mESCs and TT-SLAM-seq data upon 0 h, 2 h, 6 h and 24 h dTAG-13 treatment at the Klf4 locus. Chr, chromosome. g, FC of gene transcription (TT-SLAM-seq data) upon dTAG-13 treatment. The number of significantly deregulated genes (DESeq2) and example pluripotency genes are highlighted. h, Gene set enrichment analysis: genes are ranked according to their FC in transcription (TT-SLAM-seq) after 24 h of dTAG-13 treatment. SE genes are marked with black ticks. P denotes a nominal P value. i, Log2 FC in TT-SLAM-seq read density at SEs and typical enhancers upon dTAG-13 treatment normalized to untreated control mESCs. P values are from two-sided Wilcoxon–Mann–Whitney tests. ****P = 5 × 10⁻⁸, ***P = 5 × 10⁻⁴. j, Representative images of individual z-slices (same z) of RNA-FISH and IF signal, and an image of the merged channels. The nuclear periphery determined by DAPI staining is highlighted as a white contour (scale bars, 2.5 μm). Also shown are averaged signals of either RNA-FISH or RNAPII IF centered on the FISH foci or randomly selected nuclear positions (scale bars, 0.5 μm). r denotes a Spearman’s correlation coefficient. k, Live-cell PALM imaging of Dendra2-RNAPII and nascent RNA transcripts of Sox2-MS2 in mESCs after 24 h dTAG-13 treatment. Left, size of the nearest RNAPII cluster around Sox2; middle left, distance between the Sox2 locus and the nearest RNAPII cluster; middle right, average RNAPII cluster size globally; right, number of RNAPII clusters per cell. Data are presented as mean values ± s.d. P values are from Wilcoxon–Mann–Whitney tests.
Source data
Derepressed IAPs form nuclear foci that associate with RNAPII condensates and incorporate nearby genes
a, Representative images of individual z-slices (same z) of RNA-FISH and RNAPII IF signal, and an image of the merged channels. The nuclear periphery determined by DAPI staining is highlighted as a white contour. The zoom column displays the region of the images highlighted in a yellow box (enlarged for greater detail). Merge of the nuclear z-projections is displayed, and overlapping pixels between the RNA-FISH and IF channels are highlighted in white. Displayed MOC and Pearson’s correlation coefficient (r) values are an average obtained from 24 analyzed nuclei. Scale bars, 2.5 μm. b, Same as a, except with MED1 IF. c, Distance of IAP RNA-FISH foci to the nearest RNAPII or MED1 IF puncta. Each dot represents one IAP RNA-FISH focus. d, Meta representations of RNAPII ChIP with reference exogenous genome (ChIP-RX) (left) and MED23 ChIP–seq (right) read densities at IAP, MMERVK and MMETn ERVs in control (DMSO-) and dTAG-13 (24 h)-treated mESCs. The mean read densities are displayed ±2 kb around the indicated elements. The genomic elements were length normalized. e, Genome browser tracks at the Cthrc1 locus. Note the independent transcription initiation events at Cthrc1 and MMETn, ruling out that the MMETn acts as an alternative Cthrc1 promoter. Rpm, reads per million. f, Representative images of individual z-slices (same z) of RNA-FISH and IF signal, and an image of the merged channels. The nuclear periphery determined by DAPI staining is highlighted as a white contour (scale bars, 2.5 μm). Also shown are averaged signals of either RNA-FISH or IF centered on the Cthrc1 FISH foci or randomly selected nuclear positions (scale bars, 0.5 μm). r denotes a Spearman’s correlation coefficient. g, Same as f, except with NFYA IF. h, qRT–PCR data for IAP RNA, Cthrc1 mRNA and the Pri-miR-290-295 transcript in control and ERV-triple knockout (TKO) cells. Data are presented as mean values ± s.d. from six biological replicates. P values are from two-tailed t-tests. ****P < 1 × 10⁻⁴. i, Principal component (PC) plot of Hi-C interactions at an ERV-rich locus on chromosome 12. j, Pile-up analysis of contacts between IAPs, MMERVKs, MMETns and transcribed genes in wild-type and TRIM28-degraded mESCs.
SE-enriched TFs rescue condensate localization in TRIM28-degraded mESCs
a, Genotype of the iPSC line and scheme of the experimental setup. The iPSC line contains degradation-sensitive Trim28-FKBP alleles and doxycycline-inducible Oct4, Sox2, Klf4 and c-Myc (OSKM) transgenes. b, Western blot validation of the FKBP degron tag and OSKM ectopic expression in iPSCs. c, Representative images of IAP RNA-FISH staining. The number and percentage refer to cells with detectable IAP foci, pooled from two biological replicates. Scale bars, 10 μm; inset scale bars, 2 μm. d. Quantification of cells with detectable IAP foci (IAP⁺ cells) at the indicated treatment regimes. e, IAP RNA expression is reduced in TRIM28-degraded iPSCs that ectopically express OSKM factors. The line plot shows qRT–PCR data of IAP RNA levels normalized to 0 h of dTAG-13 treatment. Data are from three independent biological replicates (three wells on a tissue culture plate) and are presented as mean values ± s.d. The experiment was repeated three times, and data from one representative experiment are shown. P value is from two-tailed t-tests. ***P < 1 × 10⁻⁴. f, Colocalization between the nascent RNA of miR290-295 and RNAPII puncta in TRIM28-degraded iPSCs that ectopically express OSKM factors. Separate images of individual z-slices (same z) of the RNA-FISH and IF signal are shown along with an image of the merged channels. The nuclear periphery determined by DAPI staining is highlighted as a white contour (scale bars, 2.5 μm). Also shown are averaged signals of either RNA-FISH or RNAPII IF centered on the miR290-295 RNA FISH foci or randomly selected nuclear positions. r denotes a Spearman’s correlation coefficient (scale bars, 0.5 μm). g, Elevated levels of miR290-295 SE transcript and Pri-miR290-295 nascent transcript in TRIM28-degraded iPSCs that ectopically express OSKM factors. qRT–PCR data was normalized to the 0 h of dTAG-13 treatment. Data are from three independent biological replicates (three wells on a tissue culture plate) and are presented as mean values ± s.d. The experiment was repeated three times, and data from one representative experiment are shown. P values are from two-tailed t-tests. *P = 0.027, ***P = 1 × 10⁻⁴.
Source data
Contributions of IAP RNA to condensate localization in vivo and condensate formation in vitro
a, Schematic model of the ERV shRNA knockdown experiments. b, qRT–PCR data as FC-normalized to the DMSO treatment control. Data are presented as mean values ± s.d. from three biological replicates. P values are from two-tailed t-tests. ****P < 1 × 10⁻⁴, ***P < 1 × 10⁻³, **P < 1 × 10⁻². c, Log2 FC values in total RNA-seq data at intergenic SEs and genes. Data are from three biological replicates. P values are from two-sided Wilcoxon–Mann–Whitney tests. ****P < 1 × 10⁻⁴. d, Representative images of individual z-slices (same z) of RNA-FISH and IF signal, and an image of the merged channels. The nuclear periphery determined by DAPI staining is highlighted as a white contour (scale bars, 2.5 μm). Also shown are averaged signals of either RNA-FISH or RNAPII IF centered on the miR290-295 FISH foci or randomly selected nuclear positions (scale bars, 0.5 μm). r denotes a Spearman’s correlation coefficient. e, Representative images of mixtures of fluorescein-labeled IAP RNA and purified recombinant RNAPII CTD-mCherry in droplet formation buffer. Scale bar, 5 μm. f, Partitioning ratio of RNAPII CTD-mCherry into droplets at the indicated IAP RNA concentrations. Every dot represents a detected droplet. P values are from two-sided t-tests. g, Quantification of the enrichment of fluorescein-labeled IAP RNA in RNAPII CTD-mCherry droplets. P values are from two-sided t-tests. h, Quantification of the partitioning of (left) MED1 IDR and (right) HPIα into droplets in the presence of IAP RNA. Values are normalized against the partition ratio at no RNA added. Corresponding images are found in Extended Data Fig. 8a. The displayed quantification is the same as displayed in Extended Data Fig. 8b. i, Representative images of droplet formation by purified NFYC-IDR-mEGFP (1 μM) and MED1 IDR-mCherry (5 μM) fusion proteins in the presence of in vitro transcribed Cy5-labeled IAP RNA fragment. Scale bar, 5 μm. j, Partitioning ratio of NFYC-IDR-mEGFP, MED1 IDR-mCherry and IAP RNA into droplets at the indicated IAP RNA concentrations. Every dot represents a detected droplet. All pairwise P values <2.2 × 10⁻¹⁶ (Welch’s t-test). k, Schematic model of the experiment mimicking IAPEz transcription. l, qRT–PCR data as FC-normalized to the DMSO control treatment. n.d., not detectable. Data are presented as mean values ± s.d. P values are from two-tailed t-tests. ****P < 1 × 10⁻⁴. m, qRT–PCR data as FC-normalized to the DMSO control treatment. Data are presented as mean values ± s.d. from three biological replicates. P values are from two-tailed t-tests. ****P < 1 × 10⁻⁴, ***P < 1 × 10⁻³, **P < 1 × 10⁻², NS, not significant. In panels f, g, h and j, data for quantification were acquired from at least five images of two independent image series per condition.
Early ERV activation correlates with depletion of pluripotent lineages in mouse embryos
a, Scheme of the zygotic CRISPR–Cas9 perturbation platform. b, IF images of mouse E3.5 blastocysts stained for the GAG protein produced by IAPs. Nuclei are counterstained with DAPI. Note the magenta IAP GAG foci highlighted with yellow arrowheads. Scale bar, 10 µm. c, Quantification of IAP GAG foci in multiple embryos of the indicated genotype across three independent perturbation experiments. Five embryos were picked from the pool of embryos from each genotype for the staining. Each dot represents the GAG foci from an individual embryo. Data are presented as mean values ± s.d. d, Epiblast cells are depleted in TRIM28 KO embryos. Uniform manifold approximation and projection (UMAP) of E6.5 wild-type and E6.5 TRIM28 KO embryos mapped on the combined reference cell state map. The proportions of cells that belong to the individual cell states are indicated as a bar on the right of the UMAP plots. Exe, extraembryonic ectoderm. e, Lineage-specific ERV derepression in TRIM28 KO mouse embryos. The plot shows the fraction of RNA-seq reads that map to the displayed ERV taxa in the indicated cell types in wild-type (WT) and TRIM28 KO embryos in the scRNA-seq data. Each ‘x’ represents a single embryo. f, The inner part of TRIM28 KO blastocysts is populated by GATA6-expressing, NANOG-negative cells. Displayed are representative IF images of NANOG and GATA6 in E3.5 wild-type, TRIM28 KO and NANOG KO blastocysts across two independent perturbation experiments with around 20 embryos per condition. Scale bars, 20 μm. g, Condensate hijacking model. In pluripotent cells, transcriptional condensates associate with SEs bound by pluripotency TFs (for example, OCT4). In the absence of TRIM28, transcriptional condensates are lost from SEs and associate with derepressed ERVs.
Hijacking of transcriptional condensates by endogenous retroviruses

August 2022

·

339 Reads

·

45 Citations

Nature Genetics

Most endogenous retroviruses (ERVs) in mammals are incapable of retrotransposition; therefore, why ERV derepression is associated with lethality during early development has been a mystery. Here, we report that rapid and selective degradation of the heterochromatin adapter protein TRIM28 triggers dissociation of transcriptional condensates from loci encoding super-enhancer (SE)-driven pluripotency genes and their association with transcribed ERV loci in murine embryonic stem cells. Knockdown of ERV RNAs or forced expression of SE-enriched transcription factors rescued condensate localization at SEs in TRIM28-degraded cells. In a biochemical reconstitution system, ERV RNA facilitated partitioning of RNA polymerase II and the Mediator coactivator into phase-separated droplets. In TRIM28 knockout mouse embryos, single-cell RNA-seq analysis revealed specific depletion of pluripotent lineages. We propose that coding and noncoding nascent RNAs, including those produced by retrotransposons, may facilitate ‘hijacking’ of transcriptional condensates in various developmental and disease contexts.


Citations (48)


... Therefore, active DNA demethylation functions to reset or modify specific epigenetic marks that DNA methylation establishes and maintains. This process is important for processes such as embryonic development and cellular differentiation [23]. ...

Reference:

Endocrine-disrupting chemicals (EDCs) and epigenetic regulation in embryonic development: Mechanisms, impacts, and emerging trends
DNA methylation in mammalian development and disease
  • Citing Article
  • August 2024

Nature Reviews Genetics

... A central goal of developmental biology is to elucidate cell lineage relationships and molecular changes during the development of a single-celled zygote into a multicellular organism composed of numerous cell types. Most studies to date have focused on clonal tracing, which defines the descendants of a cell [5][6][7]101,102 . For example, a major question in blood development and immunology is what kinds of progenitor give rise to diverse blood and immune cell types and how their abundance changes with age. ...

Reconstructing axial progenitor field dynamics in mouse stem cell-derived embryoids
  • Citing Article
  • April 2024

Developmental Cell

... Finally, the effect of t1519 to constrain long gene expression to a low range is a novel finding. The observation may be due to a combination of distinct activating and suppressive effects of t1519 on long gene expression [4,[19][20][21]. ...

Autonomous transposons tune their sequences to ensure somatic suppression

Nature

... This is exemplified in the recent study by Hislop et al. 75 , which modeled postimplantation development without a TE compartment but succeeded in establishing yolk sac hematopoiesis, which was evident also from mapping to the embryogenesis reference (Fig. 5). Further, our tool was unable to resolve amnion cells from Pedroza et al. 72 and Karvas et al. 76 , possibly due to two distinct reasons. We observed that amnion-annotated cells from Pedroza et al. 72 are projected together with epiblast and PriS. ...

Self-patterning of human stem cells into post-implantation lineages

Nature

... In human, these methylations are associated with activation at single-copy genes 21 . H3K79me2 is enriched in euchromatin whereas H3K79me3 is enriched in repeated sequences and chromocenter 22 . In Drosophila, the level of H3K79me3 is positively correlated with gene activity 23 , which is consistent with some of the phenotypes previously reported for gpp mutants such as anterior transformations of the posterior abdominal segments 16 . ...

DOT1L bridges transcription and heterochromatin formation at mammalian pericentromeres
  • Citing Article
  • June 2023

EMBO Reports

... In elongating spermatids, the deficiency of DOT1L (a histone methyltransferase) resulted in the incomplete replacement of histones, leading to abnormal protein recruitment. This induced reduced testicular weight, sperm abnormalities, and male sterility [77,78]. Similarly, mice in which lysine-specific histone demethylase 2 (Kdm2a) was knocked out or mutated exhibit significant reductions in the total number of spermatozoa and the density of seminiferous tubules, as well as decreased sperm motility [79]. ...

DOT1L promotes spermatid differentiation by regulating expression of genes required for histone-to-protamine replacement

Development

... In most eukaryotes, methylation at the fifth position of cytosine (5mC) at CpG motifs is the most prevalent and plays a vital role in various biological processes such as gene expression regulation, genomic imprinting, transposon silencing, embryonic development, and aging [2][3][4]. CpG methylation states are not constant but can alter during development and in response to a variety of biotic and abiotic stimuli [5,6]. Consequently, precisely identifying CpG methylation states at the single-molecule level is essential to comprehensively understand and map its intricate regulatory network. ...

Dynamic antagonism between key repressive pathways maintains the placental epigenome

Nature Cell Biology

... Many viruses have been shown to form replication compartments possessing properties of BMCs, either by co-opting host proteins or encoding viral proteins that form condensates [14][15][16][17][18][19][20][21][22][23]. Recently, we demonstrated that RSV Gag forms phase-contrasted assemblies in vitro and in cells, and that these structures display properties of condensates in the nucleus, cytoplasm, and at the plasma membrane [2]. ...

Hijacking of transcriptional condensates by endogenous retroviruses

Nature Genetics

... The principal component analysis revealed that the T931del mutation impacts the transcriptomes of hemizygous males, but has little effect on that of heterozygous females (on PC3; Figs 2C, S2B and S2C). This sexual dimorphism is likely explained by the expression of the paternally inherited Ogt allele (WT) in the blastocyst [29,50], as confirmed by higher levels of Ogt transcripts in WT female blastocysts compared to WT males (S2D Fig). In good agreement, male hemizygous mutants show a more dramatic compensatory increase in Ogt expression than heterozygous mutant females (S2D Fig). ...

Diverse epigenetic mechanisms maintain parental imprints within the embryonic and extraembryonic lineages

Developmental Cell

... Although H4 hyperacetylation is a well-established modification known to precede the histone-to-protamine exchange in multiple species, other modifications, such as diand trimethylated H3K79, catalyzed by DOT1L, have been reported to temporally overlap with H4 hyperacetylation in both human and mouse spermatids (49,50). H3K79me3 is enriched at the chromocenter (the constitutive heterochromatin) of round spermatids and at repetitive elements in mESCs, whereas H3K79me2 accumulates at euchromatic regions, often downstream of promoters of actively transcribed genes (51)(52)(53)(54)(55). DOT1L loss of function mutants are embryonic lethal (56), therefore preventing the analysis of H3K79 methylation in the histone-to-protamine exchange or spermatid-specific cellular functions. ...

DOT1L bridges transcription and heterochromatin formation at pericentromeres