Integrated Transcriptome and Network Analysis Reveals Spatiotemporal Dynamics of Calvarial Suturogenesis

Greg Holmes, Ana S Gonzalez-Reiche, Na Lu, Xianxiao Zhou, Joshua Rivera, Divya Kriti, Robert Sebra, Anthony A Williams, Michael J Donovan, S Steven Potter, Dalila Pinto, Bin Zhang, Harm van Bakel, Ethylin Wang Jabs, Greg Holmes, Ana S Gonzalez-Reiche, Na Lu, Xianxiao Zhou, Joshua Rivera, Divya Kriti, Robert Sebra, Anthony A Williams, Michael J Donovan, S Steven Potter, Dalila Pinto, Bin Zhang, Harm van Bakel, Ethylin Wang Jabs

Abstract

Craniofacial abnormalities often involve sutures, the growth centers of the skull. To characterize the organization and processes governing their development, we profile the murine frontal suture, a model for sutural growth and fusion, at the tissue- and single-cell level on embryonic days (E)16.5 and E18.5. For the wild-type suture, bulk RNA sequencing (RNA-seq) analysis identifies mesenchyme-, osteogenic front-, and stage-enriched genes and biological processes, as well as alternative splicing events modifying the extracellular matrix. Single-cell RNA-seq analysis distinguishes multiple subpopulations, of which five define a mesenchyme-osteoblast differentiation trajectory and show variation along the anteroposterior axis. Similar analyses of in vivo mouse models of impaired frontal suturogenesis in Saethre-Chotzen and Apert syndromes, Twist1+/- and Fgfr2+/S252W, demonstrate distinct transcriptional changes involving angiogenesis and ribogenesis, respectively. Co-expression network analysis reveals gene expression modules from which we validate key driver genes regulating osteoblast differentiation. Our study provides a global approach to gain insights into suturogenesis.

Keywords: Fgfr2; Twist1; bone; craniofacial; craniosynostosis; differential gene expression; frontal suture; mesenchyme; metopic suture; single-cell RNA-seq.

Conflict of interest statement

Declaration of Interests The authors declare no competing interests.

Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. Regional and Temporal Gene Expression…
Figure 1.. Regional and Temporal Gene Expression Differences between SM and OFs of the FS
(A) LCM of the FS at E18.5. Section planes through frontal bones (F) and suture (S) at anterior, middle, and posterior suture levels are indicated as black lines on an ALPL-stained mouse head. SM and OF regions before and after LCM are shown on cresyl violet-stained sections. Bottom schematic indicates position of unmineralized osteoid and mineralized bone. Scale bar: 200 μm. (B) Hierarchical clustering of average expression changes for 4,282 genes (rows) with significantly increased (red) or decreased (blue) expression (FDR ≤ 0.01) in the OF compared to SM at E16.5 and E18.5 (columns). Color bar indicates the average log2 fold-change (FC). (C) Overlaps (top) and GO enrichment (bottom) of SM DEGs at E16.5 and E18.5 (p % 0.05, dashed line). (D) Overlaps (top) and GO enrichment (bottom) of OF DEGs at E16.5 and E18.5 (p ≤ 0.05, dashed line). (E) Heatmap of average expression changes between regions and stages (left) and GO BP categories (right) for 12 SM or OF genes selected for ISH validation. Significant expression changes are asterisked. Associated GO BP categories are indicated in green. Gene names are at left. (F) RNA ISH (blue) for the DEGs from (E) in the E18.5 suture, counterstained with nuclear fast red. Arrows indicate bone edges visualized by overlaid autofluorescence (yellow green). Bottom row shows bone autofluorescence (left, green) corresponding to mineralized bone stained with von Kossa (middle) as seen in merged images of adjacent sections (right). Scale bar: 200 μm. (G) Hierarchical clustering of average expression changes for 2,480 genes with significantly increased (red) or decreased (blue) expression at E18.5 compared to E16.5 in the SM and OFs (FDR ≤ 0.01). (H) Overlaps (top) and GO enrichment (bottom) of E16.5 DEGs by region (p ≤ 0.05, dashed line). (I) Overlaps (top) and GO enrichment (bottom) of E18.5 DEGs by region (p ≤ 0.05, dashed line). (J) Relative expression (2−ΔCT) by qRT-PCR for the indicated stage-specific DEGs by region. Data are shown as individual measurements from biological replicates with mean ± SD. *p % 0.05 (Student’s t test). See also Figure S1 and Table S1.
Figure 2.. Differential Splicing in the FS
Figure 2.. Differential Splicing in the FS
(A) Heatmap of the maximum percent spliced-in (PSI) changes for 261 intron clusters with significant (FDR ≤ 0.05) SM and OF regional and E16.5 and E18.5 temporal differential splicing (DS) identified by LeafCutter. (B) Proportional breakdown of event types for regional and temporal DS intron clusters (A5SS, alternative 5′ [donor] splice site; A3SS, alternative 3′ [acceptor] splice site). (C) Overlaps (top) and top GO enrichment categories (bottom; BP, biological process; MF, molecular function; CC, cellular component) for 238 genes with DS intron clusters between regions (orange) or stages (green). (D) Overview of significant DS intron clusters in Col5a1 associated with increased expression of exon E64A and decrease of the mutually exclusive exon 64B in SM compared to OFs at E16.5 and E18.5 (TSPN, thrombospondin N-terminal-like domain; COLFI, fibrillar collagen C-terminal domain; Laminin_G, laminin G domain; Collagen, collagen triple helix repeat [20 copies] domain). Increased intron usage in SM or OF is indicated in purple and green, respectively. FDR-corrected p values are indicated for each cluster for E16.5 (turquoise) and E18.5 (salmon). Details of the DS event indicating the PSI for each intron in SM and OF and the changes in PSI (DPSI) between regions for both stages. The p values for DS are indicated above each intron cluster. (E) Relative expression (2−ΔCT) by qRT-PCR confirming PSI changes at Col5a1 exons E64A and E64B (clusters 3107 and 3108) in SM and OF at E18.5. Data are shown as individual measurements from biological replicates with mean ± SD. (F) Similar to (D), but for the Postn gene (FAS1, fasciclin-like domain; TGFBI/POSTN, transforming growth factor-beta-induced protein/periostin domain). The p values for DS are indicated above each intron cluster. (G) Similar to (E), but confirming PSI changes at Postn exon E17 (cluster 2561; E17I, included; E17S, skipped). See also Figure S2 and Table S2.
Figure 3.. scRNA-Seq Analysis of the FS…
Figure 3.. scRNA-Seq Analysis of the FS at E18.5
(A) Uniform Manifold Approximation and Projection (UMAP) plot of cell-type clusters detected by unsupervised graph clustering of 6,632 cells from both replicates at E16.5 and E18.5. Related clusters of FS1–5, OB, HD, and DM cell types are outlined with a dashed line. Gene signatures used to identify cell types are in Table S3. (B) Aggregate proportion of cell types in each stage. n = 1,450 cells for E16.5 and 5,182 cells for E18.5. (C) Cell-type enrichment among genes with SM- or OF-associated expression at E16.5 or E18.5 in bulk RNA-seq data (Figure 1B). Significantly enriched cell types are asterisked (FDR ≤ 0.05). (D) Significant GO BP categories of FS1–FS5, OB, HD, and DM expression signatures (p % 0.05, Fisher’s exact test). (E) Average expression of the top five differentially expressed marker genes (boxed) ranked by FC (FDR ≤ 0.01, lnFC ≥ 0.1) for FS1–FS5, OB, HD, and DM. Genes selected for smFISH in (F) are colored according to their cluster membership. The complete list of cluster markers is in Table S3. (F) smFISH of FS1–FS5, OB, HD, and DM for the indicated genes in the E18.5 suture. OFs and adjacent bone are indicated by dashed lines, where identical shapes are used for images from the same section. The merged image at lower left is a composite of channels from two adjacent sections. The merged image at lower right is of channels from a single section. Expression is summarized in the schematic at bottom, with FS5 omitted. Scale bar: 100 μm. (G) Graph abstraction of the relationships among FS1–FS5, OB, HD, and DM. Cells are colored according to their cluster membership (left) or by pseudotime (right). A principal graph was fitted (solid and dashed lines) and rooted on the central FS1 subpopulation (circle) to determine the position of cells along an arbitrary pseudotime scale. See also Figure S3 and Table S3.
Figure 4.. Transcriptional Profiles of the FS…
Figure 4.. Transcriptional Profiles of the FS in Twist1+/− and Fgfr2+/S252W Mouse Models of Suture Dysgenesis
(A) Morphology of E18.5 WT, Twist1+/−, and Fgfr2+/S252W FSs at anterior, middle, and posterior locations (see Figure 1A for locations). Frontal bones, including OFs, are stained for ALPL (red) and nuclei (blue). Arrowheads indicate edge of OF. Wormian bones in anterior sections are asterisked. Scale bar: 100 μm. (B) FS widths at E18.5 for WT, Twist1+/−, and Fgfr2+/S252W. Data are shown as mean ± SD. *p < 0.05 (Student’s t test). (C) Hierarchical clustering of average expression changes of genes with significantly increased (red) or decreased (blue) expression in Twist1+/− or Fgfr2+/S252W FSs compared to WT in bulk RNA-seq data of SM and OF regions. Expression changes are shown as the log2 FC in expression between mutant and WT genotypes for each region and stage. Corresponding FDR-corrected p values are shown on the left as a heatmap, where shades of yellow represent significant changes. (D) Cell-type enrichment of mutant expression signatures in bulk RNA-seq data. Significantly enriched cell types are asterisked (FDR ≤ 0.05). (E) Venn diagrams showing the overlaps of significant DEGs between each mutant compared to WT in bulk and scRNA-seq datasets with corresponding hypergeometric p values indicated below each diagram. (F) Similar to (E), but showing the overlaps of significant DEGs between bulk and scRNA-seq datasets for each mutant compared to WT. (G) GO categories significantly enriched (p % 0.05; green) for DEGs between Twist1+/− and WT in bulk RNA-seq at E16.5 or E18.5 in SM or OF (top row; bulk), and per cell type at E18.5 (bottom panel of rows; single-cell). Categories are ordered from left to right by decreasing significance of enrichment among DEGs in the bulk dataset. (H) Similar to (G) but for DEGs between Fgfr2+/S252W and WT. (I) qRT-PCR for the indicated Twist1+/− and Fgfr2+/S252W DEGs in SM at E18.5. Data are shown as individual measurements from biological replicates with mean ± SD. *p % 0.05 (Student’s t test). See also Figure S4 and Tables S1 and S4.
Figure 5.. Co-expression Network Analysis of the…
Figure 5.. Co-expression Network Analysis of the SM
(A) Sunburst plot of the MEGENA co-expression network hierarchy of the SM based on aggregated bulk expression data from WT, Twist1+/−, and Fgfr2+/S252W mice. The network hierarchy is represented as concentric rings where the center ring corresponds to the root modules, which further subdivide into increasingly smaller child modules in the outward rings. Modules are colored according to the most significantly enriched GO BP category (FDR % 0.05). The network hierarchy branch for module SM286 is outlined and indicated by an arrow. (B) Similar to (A), but with modules colored according to bulk differential gene expression between region, stage, and/or genotype. (C) Similar to (A), but with modules colored according to cell-type enrichment. (D) Cell-type enrichment of the module SM286 genes. Significantly enriched cell types are asterisked (FDR ≤ 0.05). (E) Co-expression network module SM286. Genes with significantly increased expression in SM or OF are shown in red or blue, respectively. Genes with altered expression in Twist1+/− and Fgfr2+/S252W (F) or tested in in vitro OB differentiation assays (G) are shown in bold. (F) Heatmap of 24 genes in module SM286 with significant expression differences (FDR ≤ 0.05) between Twist1+/− or Fgfr2+/S252W and WT, in SM or OF regions at E16.5 or E18.5 (bulk), or per cell type at E18.5 (single-cell). Expression changes are shown as the log2 FC in expression between mutant and WT. Significant DEGs are asterisked. (G) MC3T3-E1 cells overexpressing mCherry (control), Lgr5, or Cpxm2 were differentiated for 28 days in OB differentiation media and stained with alizarin red (left) or von Kossa (right) to identify mineralized modules. (H) Expression of Lgr5 and Cpxm2 relative to Actb (×100) in MC3T3-E1 cells transfected with lentivirus expressing mCherry, Lgr5, or Cpxm2 (F) as indicated, determined by qRT-PCR. Lgr5 and Cpxm2 were overexpressed compared to mCherry control levels (*: Lgr5, p = 2.2 3 10 4; Cpxm2, p = 6.5 3 10 9; Student’s t test). See also Figure S5 and Table S5.

References

    1. Adam M, Potter AS, and Potter SS (2017). Psychrophilic proteases dramatically reduce single-cell RNA-seq artifacts: a molecular atlas of kidney development. Development 144, 3625–3632.
    1. Al-Rekabi Z, Wheeler MM, Leonard A, Fura AM, Juhlin I, Frazar C, Smith JD, Park SS, Gustafson JA, Clarke CM, et al. (2016). Activation of the IGF1 pathway mediates changes in cellular contractility and motility in single-suture craniosynostosis. J. Cell Sci 129, 483–491.
    1. Aslan H, Ravid-Amir O, Clancy BM, Rezvankhah S, Pittman D, Pelled G, Turgeman G, Zilberman Y, Gazit Z, Hoffmann A, et al. (2006). Advanced molecular profiling in vivo detects novel function of dickkopf-3 in the regulation of bone formation. J. Bone Miner. Res 21, 1935–1945.
    1. Bean CJ, Hunt PA, Millie EA, and Hassold TJ (2001). Analysis of a mal-segregating mouse Y chromosome: evidence that the earliest cleavage divisions of the mammalian embryo are non-disjunction-prone. Hum. Mol. Genet 10, 963–972.
    1. Behr B, Longaker MT, and Quarto N (2011). Craniosynostosis of coronal suture in twist1 mice occurs through endochondral ossification recapitulating the physiological closure of posterior frontal suture. Front. Physiol 2, 37.
    1. Bengtsson T, Aszodi A, Nicolae C, Hunziker EB, Lundgren-Akerlund E, and Fässler R (2005). Loss of alpha10beta1 integrin expression leads to moderate dysfunction of growth plate chondrocytes. J. Cell Sci 118, 929–936.
    1. Bentmann A, Kawelke N, Moss D, Zentgraf H, Bala Y, Berger I, Gasser JA, and Nakchbandi IA (2010). Circulating fibronectin affects bone matrix, whereas osteoblast fibronectin modulates osteoblast function. J. Bone Miner. Res 25, 706–715.
    1. Bonnet N, Garnero P, and Ferrari S (2016). Periostin action in bone. Mol. Cell. Endocrinol 432, 75–82.
    1. Brinkley JF, Fisher S, Harris MP, Holmes G, Hooper JE, Jabs EW, Jones KL, Kesselman C, Klein OD, Maas RL, et al.; FaceBase Consortium (2016). The FaceBase Consortium: a comprehensive resource for craniofacial researchers. Development 143, 2677–2688.
    1. Britanova O, Depew MJ, Schwark M, Thomas BL, Miletich I, Sharpe P, and Tarabykin V (2006). Satb2 haploinsufficiency phenocopies 2q32-q33 deletions, whereas loss suggests a fundamental role in the coordination of jaw development. Am. J. Hum. Genet 79, 668–678.
    1. Buja A, and Eyuboglu N (1992). Remarks on parallel analysis. Multivariate Behav. Res 27, 509–540.
    1. Butler A, Hoffman P, Smibert P, Papalexi E, and Satija R (2018). Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol 36, 411–420.
    1. Cao J, Spielmann M, Qiu X, Huang X, Ibrahim DM, Hill AJ, Zhang F, Mundlos S, Christiansen L, Steemers FJ, et al. (2019). The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502.
    1. Chen ZF, and Behringer RR (1995). twist is required in head mesenchyme for cranial neural tube morphogenesis. Genes Dev 9, 686–699.
    1. Christopoulos DT (2016). On the efficient identification of an inflection point. Int. J. Math. Sci. Comput 6, 13–20.
    1. Connerney J, Andreeva V, Leshem Y, Muentener C, Mercado MA, and Spicer DB (2006). Twist1 dimer selection regulates cranial suture patterning and fusion. Dev. Dyn 235, 1345–1357.
    1. Connerney J, Andreeva V, Leshem Y, Mercado MA, Dowell K, Yang X, Lindner V, Friesel RE, and Spicer DB (2008). Twist1 homodimers enhance FGF responsiveness of the cranial sutures and promote suture closure. Dev. Biol 318, 323–334.
    1. Cornelissen M, Ottelander B.d., Rizopoulos D, van der Hulst R, Mink van der Molen A, van der Horst C, Delye H, van Veelen M-L, Bonsel G, and Mathijssen I (2016). Increase of prevalence of craniosynostosis.J. Craniomaxillofac. Surg 44, 1273–1279.
    1. Cullen M, Seaman S, Chaudhary A, Yang MY, Hilton MB, Logsdon D, Haines DC, Tessarollo L, and St Croix B (2009). Host-derived tumor endothelial marker 8 promotes the growth of melanoma. Cancer Res 69, 6021–6026.
    1. Dasgupta K, and Jeong J (2019). Developmental biology of the meninges. Genesis 57, e23288.
    1. Debnath S, Yallowitz AR, McCormick J, Lalani S, Zhang T, Xu R, Li N, Liu Y, Yang YS, Eiseman M, et al. (2018). Discovery of a periosteal stem cell mediating intramembranous bone formation. Nature 562, 133–139.
    1. Demitrack ES, Gifford GB, Keeley TM, Carulli AJ, VanDussen KL, Thomas D, Giordano TJ, Liu Z, Kopan R, and Samuelson LC (2015). Notch signaling regulates gastric antral LGR5 stem cell function. EMBO J 34, 2522–2536.
    1. Denisenko E, Guo BB, Jones M, Hou R, de Kock L, Lassmann T, Poppe D, Clement O, Simmons RK, Lister R, et al. (2019). Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. bioRxiv. 10.1101/832444.
    1. DeSisto J, O’Rourke R, Bonney S, Jones HE, Guimiot F, Jones KL, and Siegenthaler JA (2019). A cellular atlas of the developing meninges reveals meningeal fibroblast diversity and function. bioRxiv. 10.1101/648642.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21.
    1. Driskell RR, Lichtenberger BM, Hoste E, Kretzschmar K, Simons BD, Charalambous M, Ferron SR, Herault Y, Pavlovic G, Ferguson-Smith AC, and Watt FM (2013). Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature 504, 277–281.
    1. Duchon A, Raveau M, Chevalier C, Nalesso V, Sharp AJ, and Herault Y (2011). Identification of the translocation breakpoints in the Ts65Dn and Ts1Cje mouse lines: relevance for modeling Down syndrome. Mamm. Genome 22, 674–684.
    1. Eguchi K, Akiba Y, Akiba N, Nagasawa M, Cooper LF, and Uoshima K (2018). Insulin-like growth factor binding Protein-3 suppresses osteoblast differentiation via bone morphogenetic protein-2. Biochem. Biophys. Res. Commun 507, 465–470.
    1. el Ghouzzi V, Le Merrer M, Perrin-Schmitt F, Lajeunie E, Benit P, Renier D, Bourgeois P, Bolcato-Bellemin AL, Munnich A, and Bonaventure J (1997). Mutations of the TWIST gene in the Saethre-Chotzen syndrome. Nat. Genet 15, 42–46.
    1. Faro C, Chaoui R, Wegrzyn P, Levaillant JM, Benoit B, and Nicolaides KH (2006). Metopic suture in fetuses with Apert syndrome at 22–27 weeks of gestation. Ultrasound Obstet. Gynecol 27, 28–33.
    1. Filzmoser P, and Gschwandtner M (2015). mvoutlier: multivariate outlier detection based on robust methods (Cran R Project).
    1. Filzmoser P, Garrett RG, and Reimann C (2005). Multivariate outlier detection in exploration geochemistry. Comput. Geosci 31, 579–587.
    1. Finak G, McDavid A, Yajima M, Deng J, Gersuk V, Shalek AK, Slichter CK, Miller HW, McElrath MJ, Prlic M, et al. (2015). MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol 16, 278.
    1. Gaut L, and Duprez D (2016). Tendon development and diseases. Wiley Interdiscip. Rev. Dev. Biol 5, 5–23.
    1. Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, Burgin G, Delorey TM, Howitt MR, Katz Y, et al. (2017). A single-cell survey of the small intestinal epithelium. Nature 551, 333–339.
    1. Han X, Wang R, Zhou Y, Fei L, Sun H, Lai S, Saadatpour A, Zhou Z, Chen H, Ye F, et al. (2018). Mapping the mouse cell atlas by microwell-seq. Cell 172, 1091–1107.e17.
    1. Hennekam RCM, Allanson JE, Gorlin RJ, and Krantz ID (2010). Gorlin’s Syndromes of the Head and Neck (Oxford University Press; ).
    1. Hermann CD, Lee CSD, Gadepalli S, Lawrence KA, Richards MA, Olivares-Navarrete R, Williams JK, Schwartz Z, and Boyan BD (2012). Interrelationship of cranial suture fusion, basicranial development, and resynostosis following suturectomy in twist1(+/-) mice, a murine model of Saethre-Chotzen syndrome. Calcif. Tissue Int 91, 255–266.
    1. Herring SW (2008). Mechanical influences on suture development and patency. Front. Oral Biol 12, 41–56.
    1. Heuzé Y, Holmes G, Peter I, Richtsmeier JT, and Jabs EW (2014). Closing the gap: genetic and genomic continuum from syndromic to nonsyndromic craniosynostoses. Curr. Genet. Med. Rep 2, 135–145.
    1. Hirukawa K, Miyazawa K, Maeda H, Kameyama Y, Goto S, and Togari A (2005). Effect of tensile force on the expression of IGF-I and IGF-I receptor in the organ-cultured rat cranial suture. Arch. Oral Biol 50, 367–372.
    1. Holleville N, Quilhac A, Bontoux M, and Monsoro-Burq AH (2003). BMP signals regulate Dlx5 during early avian skull development. Dev. Biol 257, 177–189.
    1. Holmes G (2012). The role of vertebrate models in understanding craniosynostosis. Childs Nerv. Syst 28, 1471–1481.
    1. Holmes G, Gonzalez-Reiche AS, Lu N, van Bakel H, and Jabs EW (2020a). Skeletal stem cells in craniofacial bone In Encyclopedia of Bone Biology, Zaidi M, ed. (Academic Press; ).
    1. Holmes G, van Bakel H, and Jabs EW (2020b). Transcriptome Atlases of the Craniofacial Sutures. FaceBase Consortium. .
    1. Howard TD, Paznekas WA, Green ED, Chiang LC, Ma N, Ortiz de Luna RI, Garcia Delgado C, Gonzalez-Ramos M, Kline AD, and Jabs EW (1997). Mutations in TWIST, a basic helix-loop-helix transcription factor, in Saethre-Chotzen syndrome. Nat. Genet 15, 36–41.
    1. Ikegawa M, Han H, Okamoto A, Matsui R, Tanaka M, Omi N, Miyamae M, Toguchida J, and Tashiro K (2008). Syndactyly and preaxial synpolydactyly in the single Sfrp2 deleted mutant mice. Dev. Dyn 237, 2506–2517.
    1. Iseki S, Wilkie AO, and Morriss-Kay GM (1999). Fgfr1 and Fgfr2 have distinct differentiation- and proliferation-related roles in the developing mouse skull vault. Development 126, 5611–5620.
    1. Ishii M, Merrill AE, Chan Y-S, Gitelman I, Rice DPC, Sucov HM, and Maxson RE Jr. (2003). Msx2 and Twist cooperatively control the development of the neural crest-derived skeletogenic mesenchyme of the murine skull vault. Development 130, 6131–6142.
    1. Ishii M, Sun J, Ting M-C, and Maxson RE (2015). The development of the calvarial bones and sutures and the pathophysiology of craniosynostosis. Curr. Top. Dev. Biol 115, 131–156.
    1. Izu Y, Ezura Y, Koch M, Birk DE, and Noda M (2016). Collagens VI and XII form complexes mediating osteoblast interactions during osteogenesis. Cell Tissue Res 364, 623–635.
    1. Kim HJ, Rice DP, Kettunen PJ, and Thesleff I (1998). FGF-, BMP- and Shh-mediated signalling pathways in the regulation of cranial suture morphogenesis and calvarial bone development. Development 125, 1241–1251.
    1. Klopocki E, Lohan S, Brancati F, Koll R, Brehm A, Seemann P, Dathe K, Stricker S, Hecht J, Bosse K, et al. (2011). Copy-number variations involving the IHH locus are associated with syndactyly and craniosynostosis. Am. J. Hum. Genet 88, 70–75.
    1. Koch M, Schulze J, Hansen U, Ashwodt T, Keene DR, Brunken WJ, Burgeson RE, Bruckner P, and Bruckner-Tuderman L (2004). A novel marker of tissue junctions, collagen XXII. J. Biol. Chem 279, 22514–22521.
    1. Kowalczyk MS, Tirosh I, Heckl D, Rao TN, Dixit A, Haas BJ, Schneider RK, Wagers AJ, Ebert BL, and Regev A (2015). Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res 25, 1860–1872.
    1. Kyriakides TR, Zhu YH, Smith LT, Bain SD, Yang Z, Lin MT, Danielson KG, Iozzo RV, LaMarca M, McKinney CE, et al. (1998). Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis. J. Cell Biol 140, 419–430.
    1. Lajeunie E, Le Merrer M, Arnaud E, Marchac D, and Renier D (1998). [Trigonocephaly: isolated, associated and syndromic forms. Genetic study in a series of 278 patients]. Arch. Pediatr 5, 873–879.
    1. Lakso M, Pichel JG, Gorman JR, Sauer B, Okamoto Y, Lee E, Alt FW, and Westphal H (1996). Efficient in vivo manipulation of mouse genomic sequences at the zygote stage. Proc. Natl. Acad. Sci. USA 93, 5860–5865.
    1. Law CW, Chen Y, Shi W, and Smyth GK (2014). voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol 15, R29.
    1. Lee JTH, Patikas N, Kiselev VY, and Hemberg M (2019a). Fast searches of large collections of single cell data using scfind. bioRxiv. 10.1101/788596.
    1. Lee KKL, Stanier P, and Pauws E (2019b). Mouse models of syndromic craniosynostosis. Mol. Syndromol 10, 58–73.
    1. Leung C, Tan SH, and Barker N (2018). Recent advances in Lgr5+ stem cell research. Trends Cell Biol 28, 380–391.
    1. Li B, and Dewey CN (2011). RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323.
    1. Li X, Liu P, Liu W, Maye P, Zhang J, Zhang Y, Hurley M, Guo C, Boskey A, Sun L, et al. (2005). Dkk2 has a role in terminal osteoblast differentiation and mineralized matrix formation. Nat. Genet 37, 945–952.
    1. Li J, Jin D, Fu S, Mei G, Zhou J, Lei L, Yu B, and Wang G (2013). Insulin-like growth factor binding protein-3 modulates osteoblast differentiation via interaction with vitamin D receptor. Biochem. Biophys. Res. Commun 436, 632–637.
    1. Li YI, Knowles DA, Humphrey J, Barbeira AN, Dickinson SP, Im HK, and Pritchard JK (2018). Annotation-free quantification of RNA splicing using LeafCutter. Nat. Genet 50, 151–158.
    1. Liao Y, Smyth GK, and Shi W (2014). featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930.
    1. Malfait F, and De Paepe A (2014). The Ehlers-Danlos syndrome. Adv. Exp. Med. Biol 802, 129–143.
    1. Manzanares MC, Goret-Nicaise M, and Dhem A (1988). Metopic sutural closure in the human skull. J. Anat 161, 203–215.
    1. Martin M (2011). Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J 17, 10–12.
    1. Maruyama T, Jeong J, Sheu T-J, and Hsu W (2016). Stem cells of the suture mesenchyme in craniofacial bone development, repair and regeneration. Nat. Commun 7, 10526.
    1. Mayer C, Hafemeister C, Bandler RC, Machold R, Batista Brito R, Jaglin X, Allaway K, Butler A, Fishell G, and Satija R (2018). Developmental diversification of cortical inhibitory interneurons. Nature 555, 457–462.
    1. McInnes L, Healy J, and Melville J (2018). UMAP: Uniform Manifold Approximation and Projection for dimension reduction. arXiv:1802.03426. .
    1. Merkuri F, and Fish JL (2019). Developmental processes regulate craniofacial variation in disease and evolution. Genesis 57, e23249.
    1. Miao D, and Scutt A (2002). Histochemical localization of alkaline phosphatase activity in decalcified bone and cartilage. J. Histochem. Cytochem 50, 333–340.
    1. Mironchik Y, Winnard PT Jr., Vesuna F, Kato Y, Wildes F, Pathak AP, Kominsky S, Artemov D, Bhujwalla Z, Van Diest P, et al. (2005). Twist overexpression induces in vivo angiogenesis and correlates with chromosomal instability in breast cancer. Cancer Res 65, 10801–10809.
    1. Mitchell AL, Judis LM, Schwarze U, Vaynshtok PM, Drumm ML, and Byers PH (2012). Characterization of tissue-specific and developmentally regulated alternative splicing of exon 64 in the COL5A1 gene. Connect. Tissue Res 53, 267–276.
    1. Morita H, Mazerbourg S, Bouley DM, Luo C-W, Kawamura K, Kuwabara Y, Baribault H, Tian H, and Hsueh AJW (2004). Neonatal lethality of LGR5 null mice is associated with ankyloglossia and gastrointestinal distension. Mol. Cell. Biol 24, 9736–9743.
    1. Morriss-Kay GM, and Wilkie AOM (2005). Growth of the normal skull vault and its alteration in craniosynostosis: insights from human genetics and experimental studies. J. Anat 207, 637–653.
    1. Moss ML (1954). Growth of the calvaria in the rat; the determination of osseous morphology. Am. J. Anat 94, 333–361.
    1. Motch Perrine SM, Wu M, Stephens NB, Kriti D, van Bakel H, Jabs EW, and Richtsmeier JT (2019). Mandibular dysmorphology due to abnormal embryonic osteogenesis in FGFR2-related craniosynostosis mice. Dis. Model. Mech 12, dmm038513.
    1. Nakama T, Yoshida S, Ishikawa K, Kobayashi Y, Abe T, Kiyonari H, Shioi G, Katsuragi N, Ishibashi T, Morishita R, and Taniyama Y (2016). Different roles played by periostin splice variants in retinal neovascularization. Exp. Eye Res 153, 133–140.
    1. Neben CL, Idoni B, Salva JE, Tuzon CT, Rice JC, Krakow D, and Merrill AE (2014). Bent bone dysplasia syndrome reveals nucleolar activity for FGFR2 in ribosomal DNA transcription. Hum. Mol. Genet 23, 5659–5671.
    1. Neben CL, Lay FD, Mao X, Tuzon CT, and Merrill AE (2017a). Ribo-some biogenesis is dynamically regulated during osteoblast differentiation. Gene 612, 29–35.
    1. Neben CL, Tuzon CT, Mao X, Lay FD, and Merrill AE (2017b). FGFR2 mutations in bent bone dysplasia syndrome activate nucleolar stress and perturb cell fate determination. Hum. Mol. Genet 26, 3253–3270.
    1. Nieminen P, Morgan NV, Fenwick AL, Parmanen S, Veistinen L, Mikkola ML, van der Spek PJ, Giraud A, Judd L, Arte S, et al. (2011). Inac tivation of IL11 signaling causes craniosynostosis, delayed tooth eruption, and supernumerary teeth. Am. J. Hum. Genet 89, 67–81.
    1. O’Flanagan CH, Campbell KR, Zhang AW, Kabeer F, Lim JLP, Biele J, Eirew P, Lai D, McPherson A, Kong E, et al.; CRUK IMAXT Grand Challenge Team (2019). Dissociation of solid tumor tissues with cold active protease for single-cell RNA-seq minimizes conserved collagenase-associated stress responses. Genome Biol 20, 210.
    1. Ohbayashi N, Shibayama M, Kurotaki Y, Imanishi M, Fujimori T, Itoh N, and Takada S (2002). FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis. Genes Dev 16, 870–879.
    1. Opperman LA (2000). Cranial sutures as intramembranous bone growth sites. Dev. Dyn 219, 472–485.
    1. Paladin L, Tosatto SCE, and Minervini G (2015). Structural in silico dissection of the collagen V interactome to identify genotype-phenotype correlations in classic Ehlers-Danlos Syndrome (EDS). FEBS Lett 589, 3871–3878.
    1. Park WJ, Theda C, Maestri NE, Meyers GA, Fryburg JS, Dufresne C, Cohen MM Jr., and Jabs EW (1995). Analysis of phenotypic features and FGFR2 mutations in Apert syndrome. Am. J. Hum. Genet 57, 321–328.
    1. Percival CJ, and Richtsmeier JT (2013). Angiogenesis and intramembranous osteogenesis. Dev. Dyn 242, 909–922.
    1. Philippeos C, Telerman SB, Oulès B, Pisco AO, Shaw TJ, Elgueta R, Lombardi G, Driskell RR, Soldin M, Lynch MD, and Watt FM (2018). Spatial and single-cell transcriptional profiling identifies functionally distinct human dermal fibroblast subpopulations. J. Invest. Dermatol 138, 811–825.
    1. Pritchard JJ, Scott JH, and Girgis FG (1956). The structure and development of cranial and facial sutures. J. Anat 90, 73–86.
    1. Reimand J, Kull M, Peterson H, Hansen J, and Vilo J (2007). g:Profiler–a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res 35, W193–200.
    1. Richtsmeier JT, and Flaherty K (2013). Hand in glove: brain and skull in development and dysmorphogenesis. Acta Neuropathol 125, 469–489.
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, and Smyth GK (2015). limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43, e47.
    1. Rodrigues CO, Nerlick ST, White EL, Cleveland JL, and King ML (2008). A Myc-Slug (Snail2)/Twist regulatory circuit directs vascular development. Development 135, 1903–1911.
    1. Sahar DE, Longaker MT, and Quarto N (2005). Sox9 neural crest determinant gene controls patterning and closure of the posterior frontal cranial suture. Dev. Biol 280, 344–361.
    1. Sennett R, Wang Z, Rezza A, Grisanti L, Roitershtein N, Sicchio C, Mok KW, Heitman NJ, Clavel C, Ma’ayan A, and Rendl M (2015). An integrated transcriptome atlas of embryonic hair follicle progenitors, their niche, and the developing skin. Dev. Cell 34, 577–591.
    1. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, and Ideker T (2003). Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13, 2498–2504.
    1. Song W-M, and Zhang B (2015). Multiscale embedded gene co-expression network analysis. PLoS Comput. Biol 11, e1004574.
    1. Stamper BD, Mecham B, Park SS, Wilkerson H, Farin FM, Beyer RP, Bammler TK, Mangravite LM, and Cunningham ML (2012). Transcriptome correlation analysis identifies two unique craniosynostosis subtypes associated with IRS1 activation. Physiol. Genomics 44, 1154–1163.
    1. Stempien-Otero A, Kim D-H, and Davis J (2016). Molecular networks underlying myofibroblast fate and fibrosis. J. Mol. Cell. Cardiol 97, 153–161.
    1. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, Hao Y, Stoeckius M, Smibert P, and Satija R (2019). Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, and Mesirov JP (2005). Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550.
    1. Takeshita N, Hasegawa M, Sasaki K, Seki D, Seiryu M, Miyashita S, Takano I, Oyanagi T, Miyajima Y, and Takano-Yamamoto T (2017). In vivo expression and regulation of genes associated with vascularization during early response of sutures to tensile force. J. Bone Miner. Metab 35, 40–51.
    1. Thompson EM, Baraitser M, and Hayward RD (1984). Parietal foramina in Saethre-Chotzen syndrome. J. Med. Genet 21, 369–372.
    1. Twigg SRF, and Wilkie AOM (2015). A genetic-pathophysiological framework for craniosynostosis. Am. J. Hum. Genet 97, 359–377.
    1. van den Brink SC, Sage F, Vértesy Á, Spanjaard B, Peterson-Maduro J, Baron CS, Robin C, and van Oudenaarden A (2017). Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Methods 14, 935–936.
    1. Vissers LELM, Cox TC, Maga AM, Short KM, Wiradjaja F, Janssen IM, Jehee F, Bertola D, Liu J, Yagnik G, et al. (2011). Heterozygous mutations of FREM1 are associated with an increased risk of isolated metopic craniosynostosis in humans and mice. PLoS Genet 7, e1002278.
    1. Wang Y, Xiao R, Yang F, Karim BO, Iacovelli AJ, Cai J, Lerner CP, Richtsmeier JT, Leszl JM, Hill CA, et al. (2005). Abnormalities in cartilage and bone development in the Apert syndrome FGFR2(+/S252W) mouse. Development 132, 3537–3548.
    1. Wang Y, Sun M, Uhlhorn VL, Zhou X, Peter I, Martinez-Abadias N, Hill CA, Percival CJ, Richtsmeier JT, Huso DL, and Jabs EW (2010). Activation of p38 MAPK pathway in the skull abnormalities of Apert syndrome Fgfr2(+P253R) mice. BMC Dev. Biol 10, 22.
    1. Wilk K, Yeh SA, Mortensen LJ, Ghaffarigarakani S, Lombardo CM, Bassir SH, Aldawood ZA, Lin CP, and Intini G (2017). Postnatal calvarial skeletal stem cells expressing PRX1 reside exclusively in the calvarial sutures and are required for bone regeneration. Stem Cell Reports 8, 933–946.
    1. Wilkie AO, Slaney SF, Oldridge M, Poole MD, Ashworth GJ, Hockley AD, Hayward RD, David DJ, Pulleyn LJ, Rutland P, et al. (1995). Apert syndrome results from localized mutations of FGFR2 and is allelic with Crouzon syndrome. Nat. Genet 9, 165–172.
    1. Wilkinson DG (1998). In Situ Hybridization: A Practical Approach (Oxford University Press; ).
    1. Wu M, Kriti D, van Bakel H, Jabs EW, and Holmes G (2019). Laser capture microdissection of mouse embryonic cartilage and bone for gene expression analysis. J. Vis. Exp 154 10.3791/60503.
    1. Xu PX, Adams J, Peters H, Brown MC, Heaney S, and Maas R (1999). Eya1-deficient mice lack ears and kidneys and show abnormal apoptosis of organ primordia. Nat. Genet 23, 113–117.
    1. Yin C, Zhang Y, Hu L, Tian Y, Chen Z, Li D, Zhao F, Su P, Ma X, Zhang G, et al. (2018). Mechanical unloading reduces microtubule actin crosslinking factor 1 expression to inhibit b-catenin signaling and osteoblast proliferation. J. Cell. Physiol 233, 5405–5419.
    1. Yoshida T, Vivatbutsiri P, Morriss-Kay G, Saga Y, and Iseki S (2008). Cell lineage in mammalian craniofacial mesenchyme. Mech. Dev 125, 797–808.
    1. Young ID, and Swift PG (1985). Parietal foramina in the Saethre-Chotzen syndrome. J. Med. Genet 22, 413–414.
    1. Zhang M, Faugere M-C, Malluche H, Rosen CJ, Chernausek SD, and Clemens TL (2003). Paracrine overexpression of IGFBP-4 in osteoblasts of transgenic mice decreases bone turnover and causes global growth retardation. J. Bone Miner. Res 18, 836–843.
    1. Zhang Z, Sui P, Dong A, Hassell J, Cserjesi P, Chen Y-T, Behringer RR, and Sun X (2010). Preaxial polydactyly: interactions among ETV, TWIST1 and HAND2 control anterior-posterior patterning of the limb. Development 137, 3417–3426.
    1. Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T, Tran L, Dobrin R, et al. (2013). Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153, 707–720.
    1. Zhao H, Feng J, Ho TV, Grimes W, Urata M, and Chai Y (2015). The suture provides a niche for mesenchymal stem cells of craniofacial bones. Nat. Cell Biol 17, 386–396.
    1. Zimmerman H, Yin Z, Zou F, and Everett ET (2019). Interfrontal bone among inbred strains of mice and QTL mapping. Front. Genet 10, 291.

Source: PubMed

3
Sottoscrivi