Single-cell analysis identifies a key role for Hhip in murine coronal suture development

Greg Holmes, Ana S Gonzalez-Reiche, Madrikha Saturne, Susan M Motch Perrine, Xianxiao Zhou, Ana C Borges, Bhavana Shewale, Joan T Richtsmeier, Bin Zhang, Harm van Bakel, Ethylin Wang Jabs, Greg Holmes, Ana S Gonzalez-Reiche, Madrikha Saturne, Susan M Motch Perrine, Xianxiao Zhou, Ana C Borges, Bhavana Shewale, Joan T Richtsmeier, Bin Zhang, Harm van Bakel, Ethylin Wang Jabs

Abstract

Craniofacial development depends on formation and maintenance of sutures between bones of the skull. In sutures, growth occurs at osteogenic fronts along the edge of each bone, and suture mesenchyme separates adjacent bones. Here, we perform single-cell RNA-seq analysis of the embryonic, wild type murine coronal suture to define its population structure. Seven populations at E16.5 and nine at E18.5 comprise the suture mesenchyme, osteogenic cells, and associated populations. Expression of Hhip, an inhibitor of hedgehog signaling, marks a mesenchymal population distinct from those of other neurocranial sutures. Tracing of the neonatal Hhip-expressing population shows that descendant cells persist in the coronal suture and contribute to calvarial bone growth. In Hhip-/- coronal sutures at E18.5, the osteogenic fronts are closely apposed and the suture mesenchyme is depleted with increased hedgehog signaling compared to those of the wild type. Collectively, these data demonstrate that Hhip is required for normal coronal suture development.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. scRNA-seq analysis of the wild-type…
Fig. 1. scRNA-seq analysis of the wild-type coronal suture at E16.5.
a Uniform Manifold Approximation and Projection (UMAP) plot of cell clusters detected by unsupervised graph clustering of suture-specific populations (i.e., suture mesenchyme, osteoblasts) from two replicates at E16.5. b Average expression of the top five differentially expressed marker genes (boxed) ranked by fold change (FC) (FDR % 0.01, lnFC R 0.1) for suture-specific populations. Genes selected for smFISH in c are colored according to their population membership. c Localization of populations by smFISH. Each pseudo-colored panel shows an individual section hybridized with probes for the indicated genes. The schematic summarizes the spatial distribution of populations, color-coded as in smFISH. Dashed outlines indicate frontal (f) and parietal (p) bones; white horizontal lines indicate osteoid. Sections are in the transverse plane. smFISH was performed on three independent samples with similar results. Scale bar, 50 μm. d Significant GO BP categories of population-specific expression signatures. Gene Ontology enrichment was performed with ranked query and multiple testing analytical correction (p ≤ 0.05, right of dashed line).
Fig. 2. scRNA-seq analysis of the wild-type…
Fig. 2. scRNA-seq analysis of the wild-type coronal suture at E18.5.
a UMAP plot of cell clusters detected by unsupervised graph clustering of suture-specific populations (i.e., suture mesenchyme, osteoblasts) from two replicates at E18.5. b Average expression of the top five differentially expressed marker genes (boxed) ranked by FC (FDR % 0.01, lnFC R 0.1) for suture-specific populations. Genes selected for smFISH in c are colored according to their population membership. For CS8-6, Cxcl12, which is within the top ten markers of this population, was used for its known expression in dura mater. c Localization of populations by smFISH. Each pseudo-colored panel shows an individual section hybridized with probes for the indicated genes. The schematic summarizes the spatial distribution of populations, color-coded as in smFISH. Dashed outlines indicate frontal (f) and parietal (p) bones; white horizontal lines indicate osteoid. Sections are in the transverse plane. smFISH was performed on three independent samples with similar results. Scale bar, 50 μm. d Significant GO BP categories of population-specific expression signatures. Gene Ontology enrichment was performed with ranked query and multiple testing analytical correction (p ≤ 0.05, right of dashed line).
Fig. 3. Enrichment of Hhip expression in…
Fig. 3. Enrichment of Hhip expression in SM is specific to the coronal suture.
a Average bulk gene expression changes of the significant marker genes (rows) of all SM populations across the four calvarial sutures. Expression changes are shown as the log2FC between the SM and osteogenic fronts (OF) at E16.5 and E18.5, ranked by their ‘coronal specificity score’ (CS), representing the difference between the average expression across the coronal suture data and the average expression across the remaining sutures (lambdoid, sagittal, and frontal). Frontal OF (FR), parietal OF (PA), interparietal OF (IP). b Average bulk gene expression changes of the combined top ten marker genes of SM populations CS6-4 and CS8-2 across the four calvarial sutures. Expression changes are shown as in a. c Average bulk gene expression changes of key hedgehog pathway genes across the four calvarial sutures. Expression changes are shown as in a.
Fig. 4. Fate mapping of Hhip -CreERT2-expressing…
Fig. 4. Fate mapping of Hhip-CreERT2-expressing suture mesenchyme.
a Graph abstraction of the relationships between CS8-1 to CS8-9. Inset shows coloring by cluster identity. The root state of the inferred trajectory is indicated by the number 1 at the center of the cells with the earliest pseudotime. b TdTomato-expressing (tdTomato + ) cells at postnatal day (P)2 after 1 day of induction. n = 3. c TdTomato+ cells at P3 after two consecutive days of induction. n = 3. d TdTomato+ cells at P6 after five consecutive days of induction. n = 1. e TdTomato+ cells at P30 after 1 day of induction at P1. Dashed box is enlarged at right to show cluster of tdTomato+ osteoblasts within ALPL domain (dashed lines). n = 1. f TdTomato+ cells at P90 after two consecutive days of induction at P1 and P2. n = 3. g TdTomato+ cells at P90 after five consecutive days of induction between P1-P5. n = 2. White and yellow empty/solid arrowheads indicate tdTomato+ osteoblasts/osteocytes in parietal and frontal bones, respectively. f frontal bone, p parietal bone. Sections are in the sagittal plane. Scale bars, 100 μm except inset in e, 50 μm.
Fig. 5. Coronal suture dysgenesis in Hhip…
Fig. 5. Coronal suture dysgenesis in Hhip−/− mice at E16.5.
a Staining for alkaline phosphatase activity (ALPL, red) in preosteoblasts and osteoblasts for wild type (WT) and mutant (Hhip−/−) coronal sutures. f frontal bone; p parietal bone, sm suture mesenchyme. b Immunohistochemistry for RUNX2 (green). Sections are the same as shown in a. c Immunohistochemistry for SP7 (green). d Proliferation detected by EdU incorporation (green). Sections are counterstained for ALPL activity (red) and DAPI (blue). e Quantification of EdU incorporation in osteogenic fronts (OF) and suture mesenchyme (SM). WT and Hhip−/− are indicated in black and red, respectively. n = 3 WT and three Hhip−/−. f Quantification of cell numbers per section in OFs and SM. n = 3 WT and three Hhip−/−. g TUNEL assay for apoptotic cells. Inset, example of an apoptotic cell in non-sutural tissue on the same slide. White dashed outlines, frontal and parietal bones. Data in e and f are presented as dot plots showing mean ± standard deviation. Results presented in a, b, c, d, and g are representative of n = 3 WT and three Hhip−/− independent samples. Sections are in the transverse plane. Scale bar, 50 μm. Source data are provided as a Source Data file.
Fig. 6. Coronal suture dysgenesis in Hhip…
Fig. 6. Coronal suture dysgenesis in Hhip−/− mice at E18.5.
a Staining for alkaline phosphatase activity (ALPL, red) in preosteoblasts and osteoblasts for WT and Hhip−/− coronal sutures. f, frontal bone; p, parietal bone; sm, suture mesenchyme. b Immunohistochemistry for RUNX2 (green). Sections are the same as shown in a. c Immunohistochemistry for SP7 (green). d Proliferation detected by EdU incorporation (green). Sections are counterstained for ALPL activity (red) and DAPI (blue). e Quantification of EdU incorporation in osteogenic fronts (OF) and suture mesenchyme (SM). WT and Hhip−/− are indicated in black and red, respectively. Asterisk indicates a significant difference using the two-tailed Student’s t-test (P = 0.030). n = 6 WT and six Hhip−/−. f Quantification of cell numbers per section in OFs and SM. Asterisk indicates a significant difference using the two-tailed Student’s t-test (P = 0.018, 0.008, and 0.00004, respectively). n = 6 WT and six Hhip−/−. g TUNEL assay for apoptotic cells. Inset, example of an apoptotic cell in non-sutural tissue on the same slide. White dashed outlines, frontal and parietal bones. Data in e and f are presented as dot plots showing mean ± standard deviation. Results presented in a, b, c, and g are representative of n = 3 WT and three Hhip−/− independent samples. Sections are in the transverse plane. Scale bar, 100 μm. Source data are provided as a Source Data file.
Fig. 7. Results of principal components analyses…
Fig. 7. Results of principal components analyses (PCA) of data from microcomputed tomography (microCT) imaging of Hhip−/− heads at E18.5.
Scatter plots of results of PC1 and PC2 scores based on PCA analysis of unique linear distances among 3D landmarks for a form and b shape (adjusting for the effects of allometry) of WT (n = 4) and Hhip−/− (n = 7) skulls. c 3D reconstruction of microCT images showing dorsal views of the frontal (f) and parietal (p) bones of WT and Hhip−/− calvaria. Insets show lateral views of the whole skull. Images are representative n = 4 WT and seven Hhip−/− skulls. Scale bars, 1 mm.
Fig. 8. Localization of Hhip −/− coronal…
Fig. 8. Localization of Hhip−/− coronal suture populations by smFISH.
Each pseudo-colored panel shows an individual section hybridized with probes for the indicated genes at each age. Expression of the E18.5 CS8-2 marker, Cd34, can be compared to WT in Supplementary Fig. 3a. Schematics summarize the spatial distribution of populations, color-coded as in smFISH. White arrowheads indicate novel Mmp13 expression. Dashed outlines indicate frontal (f) and parietal (p) bones; white horizontal lines indicate osteoid. Sections are in the transverse plane. smFISH was performed on three independent samples with similar results. Scale bars, 50 μm.
Fig. 9. Altered HH signaling in the…
Fig. 9. Altered HH signaling in the Hhip−/− coronal suture.
a LacZ staining (blue) in WT, Hhip+/−, and Hhip−/− coronal sutures at E18.5, counterstained with nuclear fast red. be smFISH for b, dPtch1, and c, eGli1 in WT and Hhip−/− coronal sutures at b, c E16.5 and d, e E18.5. Dashed outlines indicate frontal (f) and parietal (p) bones; sm, suture mesenchyme. Sections are in the transverse plane. Results presented in a are representative of n = 2 WT, two Hhip+/−, and two Hhip−/− independent samples. Results presented in be are representative of n = 3 WT and three Hhip−/− independent samples. Scale bars, 50 μm.

References

    1. Ishii M, Sun J, Ting M-C, Maxson RE. The development of the calvarial bones and sutures and the pathophysiology of craniosynostosis. Curr. Top. Dev. Biol. 2015;115:131–156.
    1. Mundlos S, et al. Mutations involving the transcription factor CBFA1 cause cleidocranial dysplasia. Cell. 1997;89:773–779.
    1. Lee B, et al. Missense mutations abolishing DNA binding of the osteoblast-specific transcription factor OSF2/CBFA1 in cleidocranial dysplasia. Nat. Genet. 1997;16:307–310.
    1. Cohen, M. M. in Craniosynostosis: Diagnosis, Evaluation, and Management 2nd edn, Vol. 37 (eds. Cohen, M. M. & MacLean, R. E.) 112–118 (Oxford University Press, 2000).
    1. Goos JAC, Mathijssen IMJ. Genetic causes of craniosynostosis: an update. Mol. Syndromol. 2019;10:6–23.
    1. Wilkie AOM, Johnson D, Wall SA. Clinical genetics of craniosynostosis. Curr. Opin. Pediatr. 2017;29:622–628.
    1. Twigg SRF, Wilkie AOM. A genetic-pathophysiological framework for craniosynostosis. Am. J. Hum. Genet. 2015;97:359–377.
    1. Heuzé Y, Holmes G, Peter I, Richtsmeier JT, Jabs EW. Closing the gap: genetic and genomic continuum from syndromic to nonsyndromic craniosynostoses. Curr. Genet. Med. Rep. 2014;2:135–145.
    1. Jiang X, Iseki S, Maxson RE, Sucov HM, Morriss-Kay GM. Tissue origins and interactions in the mammalian skull vault. Dev. Biol. 2002;241:106–116.
    1. Yoshida T, Vivatbutsiri P, Morriss-Kay G, Saga Y, Iseki S. Cell lineage in mammalian craniofacial mesenchyme. Mech. Dev. 2008;125:797–808.
    1. Deckelbaum RA, et al. Regulation of cranial morphogenesis and cell fate at the neural crest-mesoderm boundary by engrailed 1. Development. 2012;139:1346–1358.
    1. Bernardini C, et al. Gene expression profiling in human craniosynostoses: a tool to investigate the molecular basis of suture ossification. Childs Nerv. Syst. 2012;28:1295–1300.
    1. Holmes G, et al. Integrated transcriptome and network analysis reveals spatiotemporal dynamics of calvarial suturogenesis. Cell Rep. 2020;32:107871.
    1. Chuang P-T, Kawcak T, McMahon AP. Feedback control of mammalian hedgehog signaling by the hedgehog-binding protein, Hip1, modulates Fgf signaling during branching morphogenesis of the lung. Genes Dev. 2003;17:342–347.
    1. Han X, et al. Mapping the mouse cell atlas by microwell-seq. Cell. 2018;172:1091–1107.e17.
    1. Driskell RR, et al. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature. 2013;504:277–281.
    1. Philippeos C, et al. Spatial and single-cell transcriptional profiling identifies functionally distinct human dermal fibroblast subpopulations. J. Invest. Dermatol. 2018;138:811–825.
    1. Sennett R, et al. An integrated transcriptome atlas of embryonic hair follicle progenitors, their niche, and the developing skin. Dev. Cell. 2015;34:577–591.
    1. DeSisto J, et al. Single-cell transcriptomic analyses of the developing meninges reveal meningeal fibroblast diversity and function. Dev. Cell. 2020;54:43–59.e4.
    1. Farmer DT, et al. The developing mouse coronal suture at single-cell resolution. Nat. Commun. 2021;12:4797.
    1. Pitirri MK, Kawasaki K, Richtsmeier JT. It takes two: building the vertebrate skull from chondrocranium and dermatocranium. Vertebr. Zool. 2020;70:587–600.
    1. Holmes G, et al. Early onset of craniosynostosis in an Apert mouse model reveals critical features of this pathology. Dev. Biol. 2009;328:273–284.
    1. Johnson D, Iseki S, Wilkie AO, Morriss-Kay GM. Expression patterns of Twist and Fgfr1, −2 and −3 in the developing mouse coronal suture suggest a key role for twist in suture initiation and biogenesis. Mech. Dev. 2000;91:341–345.
    1. Iseki S, Wilkie AO, Morriss-Kay GM. Fgfr1 and Fgfr2 have distinct differentiation- and proliferation-related roles in the developing mouse skull vault. Development. 1999;126:5611–5620.
    1. Holmes G. The role of vertebrate models in understanding craniosynostosis. Childs Nerv. Syst. 2012;28:1471–1481.
    1. Lee KKL, Stanier P, Pauws E. Mouse models of syndromic craniosynostosis. Mol. Syndromol. 2019;10:58–73.
    1. Brinkley JF, et al. The FaceBase consortium: a comprehensive resource for craniofacial researchers. Development. 2016;143:2677–2688.
    1. Samuels, B. D. et al. FaceBase 3: analytical tools and FAIR resources for craniofacial and dental research. Development147, dev191213 (2020).
    1. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 2020;15:1484–1506.
    1. Madisen L, et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 2010;13:133–140.
    1. Pathi S, Rutenberg JB, Johnson RL, Vortkamp A. Interaction of Ihh and BMP/Noggin signaling during cartilage differentiation. Dev. Biol. 1999;209:239–253.
    1. Doro D, Liu A, Grigoriadis AE, Liu KJ. The osteogenic potential of the neural crest lineage may contribute to craniosynostosis. Mol. Syndromol. 2019;10:48–57.
    1. Quarto N, et al. Origin matters: differences in embryonic tissue origin and Wnt signaling determine the osteogenic potential and healing capacity of frontal and parietal calvarial bones. J. Bone Miner. Res. 2010;25:1680–1694.
    1. Quarto N, Behr B, Li S, Longaker MT. Differential FGF ligands and FGF receptors expression pattern in frontal and parietal calvarial bones. Cells Tissues Organs. 2009;190:158–169.
    1. Hardy E, Fernandez-Patron C. Destroy to rebuild: the connection between bone tissue remodeling and matrix metalloproteinases. Front. Physiol. 2020;11:47.
    1. Tang SY, Herber R-P, Ho SP, Alliston T. Matrix metalloproteinase-13 is required for osteocytic perilacunar remodeling and maintains bone fracture resistance. J. Bone Miner. Res. 2012;27:1936–1950.
    1. Johansson N, et al. Collagenase-3 (MMP-13) is expressed by hypertrophic chondrocytes, periosteal cells, and osteoblasts during human fetal bone development. Dev. Dyn. 1997;208:387–397.
    1. Lana-Elola E, Rice R, Grigoriadis AE, Rice DPC. Cell fate specification during calvarial bone and suture development. Dev. Biol. 2007;311:335–346.
    1. Zhao H, et al. The suture provides a niche for mesenchymal stem cells of craniofacial bones. Nat. Cell Biol. 2015;17:386–396.
    1. Maruyama T, Jeong J, Sheu T-J, Hsu W. Stem cells of the suture mesenchyme in craniofacial bone development, repair and regeneration. Nat. Commun. 2016;7:10526.
    1. Wilk K, et al. Postnatal calvarial skeletal stem cells expressing PRX1 reside exclusively in the calvarial sutures and are required for bone regeneration. Stem Cell Rep. 2017;8:933–946.
    1. Guo Y, et al. BMP-IHH-mediated interplay between mesenchymal stem cells and osteoclasts supports calvarial bone homeostasis and repair. Bone Res. 2018;6:30.
    1. Debnath S, et al. Discovery of a periosteal stem cell mediating intramembranous bone formation. Nature. 2018;562:133–139.
    1. Yu M, et al. Cranial suture regeneration mitigates skull and neurocognitive defects in craniosynostosis. Cell. 2021;184:243–256.e18.
    1. Maruyama, T. et al. BMPR1A maintains skeletal stem cell properties in craniofacial development and craniosynostosis. Sci. Transl. Med. 13, eabb4416 (2021).
    1. Di Pietro, L. et al. GLI1 and AXIN2 are distinctive markers of human calvarial mesenchymal stromal cells in nonsyndromic craniosynostosis. Int. J. Mol. Sci. 21, 4356 (2020).
    1. Park S, Zhao H, Urata M, Chai Y. Sutures possess strong regenerative capacity for calvarial bone injury. Stem Cells Dev. 2016;25:1801–1807.
    1. Pan A, Chang L, Nguyen A, James AW. A review of hedgehog signaling in cranial bone development. Front. Physiol. 2013;4:61.
    1. Jacob S, Wu C, Freeman TA, Koyama E, Kirschner RE. Expression of Indian Hedgehog, BMP-4 and Noggin in craniosynostosis induced by fetal constraint. Ann. Plast. Surg. 2007;58:215–221.
    1. Rice DPC, et al. Gli3Xt-J/Xt-J mice exhibit lambdoid suture craniosynostosis which results from altered osteoprogenitor proliferation and differentiation. Hum. Mol. Genet. 2010;19:3457–3467.
    1. Veistinen LK, et al. Regulation of calvarial osteogenesis by concomitant de-repression of GLI3 and activation of IHH targets. Front. Physiol. 2017;8:1036.
    1. Lenton K, et al. Indian hedgehog positively regulates calvarial ossification and modulates bone morphogenetic protein signaling. Genesis. 2011;49:784–796.
    1. Abzhanov A, Rodda SJ, McMahon AP, Tabin CJ. Regulation of skeletogenic differentiation in cranial dermal bone. Development. 2007;134:3133–3144.
    1. St-Jacques B, Hammerschmidt M, McMahon AP. Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev. 1999;13:2072–2086.
    1. Klopocki E, et al. Copy-number variations involving the IHH locus are associated with syndactyly and craniosynostosis. Am. J. Hum. Genet. 2011;88:70–75.
    1. Barroso E, et al. Identification of the fourth duplication of upstream IHH regulatory elements, in a family with craniosynostosis Philadelphia type, helps to define the phenotypic characterization of these regulatory elements. Am. J. Med. Genet. A. 2015;167A:902–906.
    1. Will AJ, et al. Composition and dosage of a multipartite enhancer cluster control developmental expression of Ihh (Indian hedgehog) Nat. Genet. 2017;49:1539–1545.
    1. Feng W, Choi I, Clouthier DE, Niswander L, Williams T. The Ptch1(DL) mouse: a new model to study lambdoid craniosynostosis and basal cell nevus syndrome-associated skeletal defects. Genesis. 2013;51:677–689.
    1. Twigg SRF, et al. A recurrent mosaic mutation in SMO, encoding the hedgehog signal transducer smoothened, is the major cause of Curry-Jones syndrome. Am. J. Hum. Genet. 2016;98:1256–1265.
    1. Jenkins D, et al. RAB23 mutations in Carpenter syndrome imply an unexpected role for hedgehog signaling in cranial-suture development and obesity. Am. J. Hum. Genet. 2007;80:1162–1170.
    1. McDonald-McGinn DM, et al. Metopic craniosynostosis due to mutations in GLI3: a novel association. Am. J. Med. Genet. A. 2010;152A:1654–1660.
    1. Shi Y, et al. Gli1 identifies osteogenic progenitors for bone formation and fracture repair. Nat. Commun. 2017;8:2043.
    1. Wang Y, et al. Activation of p38 MAPK pathway in the skull abnormalities of Apert syndrome Fgfr2(+P253R) mice. BMC Dev. Biol. 2010;10:22.
    1. Bean CJ, Hunt PA, Millie EA, Hassold TJ. Analysis of a malsegregating mouse Y chromosome: evidence that the earliest cleavage divisions of the mammalian embryo are non-disjunction-prone. Hum. Mol. Genet. 2001;10:963–972.
    1. Holtwick R, et al. Smooth muscle-selective deletion of guanylyl cyclase-A prevents the acute but not chronic effects of ANP on blood pressure. Proc. Natl Acad. Sci. USA. 2002;99:7142–7147.
    1. Filzmoser, P. and Gschwandtner, M. mvoutlier: multivariate outlier detection based on robust methods. (2015).
    1. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20:296.
    1. Stuart T, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888–1902.e21.
    1. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 2018;36:411–420.
    1. Haber AL, et al. A single-cell survey of the small intestinal epithelium. Nature. 2017;551:333–339.
    1. Reimand J, Kull M, Peterson H, Hansen J, Vilo J. g:Profiler–a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res. 2007;35:W193–W200.
    1. Ritchie ME, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
    1. Cao J, et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature. 2019;566:496–502.
    1. Holmes, G. et al. Midface and upper airway dysgenesis in FGFR2-related craniosynostosis involves multiple tissue-specific and cell cycle effects. Development145, dev166488 (2018).
    1. Motch Perrine SM, et al. Craniofacial divergence by distinct prenatal growth patterns in Fgfr2 mutant mice. BMC Dev. Biol. 2014;14:8.

Source: PubMed

3
Sottoscrivi