REGENERATIVE MEDICINE

Time Point-Based Integrative Analyses of Deep- Transcriptome Identify Four Signal Pathways in Blastemal Regeneration of Zebrafish Lower Jaw

a a a a b a,c HUI ZHANG, XUELONG WANG, KAILUN LYU, SIQI GAO, GUAN WANG, CHUNXIN FAN, d a,c XIN A. ZHANG, JIZHOU YAN

Key Words. Zebrafish • Blastemal regeneration • foxi1 • RNA-sequencing • Tissue engineering aDepartment of Biology, ABSTRACT Institute for Marine There has been growing interest in applying tissue engineering to stem cell-based regeneration Biosystem and therapies. We have previously reported that zebrafish can faithfully regenerate complicated tis- Neurosciences, College of sue structures through blastemal cell type conversions and tissue reorganization. To unveil the Fisheries and Life Sciences; regenerative factors and engineering arts of blastemal regeneration, we conducted transcripto- and cKey Laboratory of mal analyses at four time points corresponding to preamputation, re-epitheliation, blastemal Exploration and Utilization of Aquatic Genetic Resources formation, and respecification. By combining the hierarchical ontology term network, the Ministry of Education, DAVID annotation system, and Euclidean distance clustering, we identified four signaling path- Shanghai Ocean University ways: foxi1-foxo1b-, pax3a-mant3a-col11/col2, pou5f1--kdrl, and -wnt11 PCP- Shanghai People’s Republic of sox9a. Results from immunohistochemical staining and promoter-driven transgenic fish suggest China; bGenergy that these pathways, respectively, define wound epidermis reconstitution, cell type conversions, Biotechnology (Shanghai) Co., blastemal angiogenesis/vasculogenesis, and cartilage matrix-orientation. Foxi1 morpholino- Ltd., Shanghai, People’s knockdown caused expansions of Foxo1b- and Pax3a-expression in the basal layer-blastemal Republic of China; junction region. Moreover, foxi1 morphants displayed increased sox9a and hoxa2b transcripts in dStephenson Cancer Center the embryonic pharyngeal arches. Thus, a Foxi1 signal switch is required to establish correct tis- and Department of sue patterns, including re-epitheliation and blastema formation. This study provides novel Physiology, The University of insight into a blastema regeneration strategy devised by epithelial cell transdifferentiation, Oklahoma Health Sciences blood vessel engineering, and cartilage matrix deposition. STEM CELLS 2015;33:806–818 Center, Oklahoma City, Oklahoma, USA

Correspondence: Jizhou Yan, M.D., INTRODUCTION tissue engineering, devising blood vessel- Ph.D., Institute for Marine engineered tissue becomes a challenge, particu- Biosystem and Neurosciences, Most invertebrate species, certain urodeles, larly when designing a completely scaffold-free Department of Biology, College of and fish can faithfully regenerate their lost tis- product [4]. One potential improvement involves Fisheries and Life Sciences, Ministry sues and appendage organs, whereas most of Education, Shanghai Ocean the delivery of constructive cues to and modula- University, Shanghai 201306, China. vertebrate species and mammals, including tion of the micro-environmental niche. Bioactive Telephone: 862161900488; Fax: humans, do not have such regenerative molecules could be used to spatially direct stem 8621-61900445; e-mail: jyan2@ capacity [1, 2]. Regenerative medicine aims to cell differentiation into multiple tissues [6]. shou.edu.cn help humans restore the lost tissues, mostly Unfortunately, no such bioactive factors or bioma- Received May 18, 2014; through stem cell-based tissue engineering. terials are well established. Thus, identifying the accepted for publication Various stem cell types, including embryonic active factors and clarifying their interrelationships October 30, 2014; first stem cells (ESCs), induced pluripotent stem in distinct tissue niches from species with high STEM CELLS published online in cells (iPS), and adult stem cells, such as bone tissue regeneration capabilities will aid in the EXPRESS November 25, 2014. marrow mesenchymal stem cells and skin stem identification and design of versatile biomaterials. VC AlphaMed Press cells or epidermal neural crest stem cells (EPI- Zebrafish can faithfully regenerate compli- 1066-5099/2014/$30.00/0 NCSC), are being investigated [3–6]. Immuno- cated tissue structures. In our previous study genicity of cell-based tissue replacement, the http://dx.doi.org/ [7], we described the regenerative process of 10.1002/stem.1899 potential for teratoma or uncontrolled cell pro- the lower jaw and determined three time liferation of ESC or iPS cells, and structural dis- points that correspond to epidermis reconstruc- organization of stem cell components limit the tion, blastema formation, and reformation. clinical translation efficacy for such regenera- Understanding the cellular and molecular path- tive strategies. ways by which fish regenerate multiple tissue Tissue engineering focuses on the construc- structures will help us to refine the regenerative tion of three-dimensional neotissues from their rules for controlling proliferation and patterning cellular components. Since angiogenesis is essen- in mammals and humans [8]. High-throughput tial in almost all types of organ remodeling and transcriptomal analysis is an ideal approach to

STEM CELLS 2015;33:806–818 www.StemCells.com VC AlphaMed Press 2014 Zhang, Wang, Lyu et al. 807 identify the molecules which orchestrate the successive cell-type flink [15] to analyze gene and transcript expressions. After the conversions, and provide regeneration mechanisms for tissue normalization of RNA read counts based on the library size, engineering or for induction of blastemal regeneration. Although sequencing depth differences, and gene length differences, we several papers have described the use of deep sequencing in conducted differential analysis for each pair of comparison animal regeneration studies [9–11], our study combined stereo- groups using Cuffdiff software [15]. The differential expression typed cellular responses with time-lapse transcriptomal integra- (cut-off value) between two transcripts was defined as greater tive analyses, and examined genotype-phenotype correlations than twofold (log 2 ratio >1) with p < .05. that occur during the course of lower jaw regeneration. Functional Annotations All differentially expressed gene and isoform sequences MATERIALS AND METHODS (q < 0.05) were annotated in the gene ontology (GO) database Zebrafish Husbandry (www.geneontology.org) and the Kyoto Encyclopedia of and Genomes (KEGG) pathway database (www.genome.jp/kegg/ Zebrafish AB lines were maintained under an ambient temper- pathway.html). To compare two time points of samples with two  ature of 27 C with a timer-controlled light cycle of 14 hours replicates, we used R package (goseq) from bioconductor light and 10 hours dark. All experiments were performed in (http://www.bioconductor.org/) to implement enrichment analy- accordance with the Animal Care and Use Committee guide- sis for GO annotation and pathways. The p values of the enrich- lines of Shanghai Ocean University. ment were calculated and adjusted to q-values for multiple testing corrections based on the hypergeometric distribution Generation of Transgenic Fishes [16]. Meanwhile, the false discovery rate was determined Zebrafish pou5f1 promoter-derived green fluorescent according to Benjamini methods [17]. The generated database (GFP) expression construct pGFP-6.5 was a gift from Professor (time point-based differential GO analysis) was used for the hier- Kyo Yamasu. Pou5f1:GFP transgenic fish lines were generated archical GO term network that we describe in the next section. as previously described [12]. Flk1:GFP and Isl1:GFP transgenic fish were gifts from Yingbing Zhong and Chunxin Fan (China Hierarchical GO Term Network Zebrafish Resource Center), respectively. To generate the GO term network, we first identified all the superfamilies in superfamily (dcGO) and created files for them Tissue Processing, Histology, and (BP_SuperFam_GO.txt, MF_SuperFam_GO.txt, and CC_Super- Immunohistochemistry Fam_GO.txt). The root of the category starts from this dcGO Histology, immunohistochemical staining, and RNA in situ (http://supfam.org/SUPERFAMILY/cgi-bin/dcgo.cgi). Then, we hybridization were performed as described (Supporting Infor- downloaded the GO database and mounted it on Oracle. mation Table S11) [7, 13]. The antibody specificity was tested Based on the superfamilies, a script was written to retrieve in a zebrafish embryonic fibroblast cell line, pac2 (Supporting the GO hierarchy down to the next level (GO_Hier_Tree.txt). Information Fig. S1). Then, a program (PerlScript) was built to locate every daugh- ter GO term. In order to avoid a recursive loop, we simply Morpholino Injections checked each of the parent GO ids and prevented them from Morpholinos were injected into the yolk sacs of embryos at appearing in the list of target daughter GO terms. Finally, we the one-cell stage, as described [14]. Procedures for morpho- used the road map GO_Hier_Tree.txt (A file) to annotate the lino tissue injection were performed as described previously [7] files of temporally differential GO analysis. with the following modifications. After amputation, morpholino was injected into the wound at three injection sites (1 mlof Functional Classification and Clustering Enrichment 1 mmol each site) and electroporated four pulses at 50 V for In order to understand the biological functions of unigene and 30 milliseconds with 5-second intervals using a BTX Electro isoforms, including the cluster of orthologous groups, we used Square Porator ECM 830 (BTX, San Diego, CA, http://www.harvar- the DAVID annotation system (http://david.abcc.ncifcrf.gov/) to dapparatus.com/). All morpholino oligonucleotides (Mo) were perform functional classification and term enrichment [18, 19]. purchased from Gene Tools, LLC (Philomath, OR, http://www. gene-tools.com/). According to the manufacturer’s recommenda- Hierarchical Clustering of Differential Expression Data tions, Vivo-Morpholino antisense oligonucleotides corresponding To determine the similarity of differentially expressed genes to zebrafish sequences of foxi1, sox9a,andhoxa2b mRNAs were between two time points, we performed hierarchical cluster- directed against the translation start sites. A nonsense standard ing. In hierarchical clustering, genes with similar expression control morpholino was used to evaluate unspecific effects due patterns are grouped together and connected by a clustering to the morpholino injection. The morpholino efficiency was eval- tree or dendrogram. The distance in clusters among genes uated at protein level by quantitative comparison of immunohis- was calculated using SAS (SAS Institute, Inc., 2011, Cary, NC). tochemical staining (Supporting Information Fig. S2) [13].

RNA Deep Sequencing Analyses RESULTS Two intervals of Illumina RNA-deep sequencing were performed as described [7]. All data are available at the NCBI short read Wound Epidermis Constitution and Blastema archive under study number SRA048162.1. All sequenced raw Formation FASTQdataweremappedonthezebrafishreferencegenome Zebrafish epidermis consists of a surface layer, an intermedi- (UCSC, danRer7) using Tophat. For each sample, we used Cuf- ate layer, and a basal layer (Supporting Information Fig. S3). www.StemCells.com VC AlphaMed Press 2014 808 Four Signal Pathways in Jaw Blastemal Regeneration

Figure 1. Re-epitheliation of the wound epidermis. Panels (A) and (B) show actual images of the intact lower jaw at the ventral side of the zebrafish head (A); and the histological structure of the intact lower jaw at transverse section and HE staining (B). The line demarcates the amputation plane in the following experiments. After amputation, blood cells (nbc) first accumulated on the wound muscle (Mm) (C, D), followed by epidermal cells and ctc from the surrounding tissues (E, F). These cells mixed with collagen matrix (col), reconstituted the wound epidermis, and underwent morphological changes toward epidermal cells. The cells with large, dark nuclei with little cytoplasm or having vacuolar cytoplasm are designated as the intermediate forms (imc) (G, H). HE staining; Scale bars (A) 5 500 mm; (B), (C), (E), (G) 5 50 mm; (D), (F), (H) 5 20 mm. Abbreviations: BM, basal membrane; ctc, connective tissue cells; Der, der- mis; Epi, epidermis; Hyp, hypodermis; kc, kerotine cells; mc, mucous cells; Md, mandible; Mm, mandibular muscle; Ms, mandibular sym- physis; nbc, nucleated blood cells; sc, small nucleated cells.

Supposedly, the intermediate layer contains various types of blood, epidermis, disrupted muscle, and associated connective differentiated and undifferentiated epidermal cells, while the tissues accumulated on the wound bed. These cells inter- basal layer is composed of undifferentiated cells and tightly weaved with collagen matrix and constituted wound epider- links epidermis to dermis [20]. After a transverse removal of mis (Fig. 1C–1F). At 24 hpa, the newly formed epithelium one-third of the mandible and associated epidermis, dermis, consisted of epidermal and intermediate cells (Fig. 1G, 1H). hypodermis, mandibular symphysis, and part of intermandibu- The intermediate cells displayed nuclear condensation and laris anterior (Fig. 1A, 1B), epidermis reconstitution was initi- cytoplasmic shrinkage or vacuolation, and were morphologi- ated following homeostasis. At least four sources of cells from cally between mesenchymal cells/nbc and epidermal

VC AlphaMed Press 2014 STEM CELLS Zhang, Wang, Lyu et al. 809

Figure 2. Cell type conversions in mesenchymal blastema and chondrogenic blastema from 2 dpa to 7 dpa. At 2 dpa, relatively undif- ferentiated bc were characterized by a high nucleus-to-cytoplasm ratio, clear nucleoli, and basophilic cytoplasm (A). With increased extracellular matrix, heterogenous blastemal cells were activated, and became fc (B,C). Panels (D)–(F) show progressive differentiation and compartmentalization of blastemal cells toward Coc and Mm. Insets show the blastemal cell morphology by magnification of the same section. Scale bars 5 100 mm. Abbreviations: bc, blastemal cells; Coc, chondrogenic center; fc, fibroblast-like cells; Mm, mandibular muscle; sc, small nucleated cell. keratinocytes/mucous cells. We attributed the complete re- the outer area of the blastema became reorganized to resem- epitheliation to cell-type conversions to tissue interactions ble the shape and polarity of the adjacent tissues. Their long between ectodermal-mesodermal components. axes were oriented approximately parallel to the long axis of As the stratified epidermis covered the entire wound and the adjoining mandible and muscle fiber (Fig. 2D–2F). We tightly adhered to the wound’s basal membrane, an aggregate intended to determine which molecules mark the cell identity, of small nucleated cells (sc) with a minimal amount of cyto- and which signaling pathways orchestrate the dynamics of cell plasm emerged in the prospective blastema, just beneath the type conversion, angiogenesis, and extracellular matrix (ECM) wound epidermis, at 1 dpa (Fig. 1H). When vasculogenesis deposition. and angiogenesis became active at 2 dpa, diverse cell types increased in number and gradually changed their morphology Deep Sequencing of Total RNAs from Four Significant toward a fibroblast-like phenotype (Fig. 2A, 2B). On the fourth Stages During Lower Jaw Regeneration day, an expanding mesenchymal blastema was clearly visible, To obtain a comprehensive non-biased view of gene activity characterized by a mass of fibroblast-like blastemal cells (bc) during different stages of jaw regeneration, we conducted a packed in the increased extracellular matrix and fibrous tis- large scale deep sequencing of RNA samples. We monitored sues (Fig. 2C). On the fifth day, the blastemal cells in the cen- the changes in gene expression at four representative stages: tral zone of the blastema were arranged circularly and uncut (0 hpa, hours postamputation), 2 hpa, 2 dpa (days post- converted into chondrogenic blastema. The cells located in amputation), and 5 dpa (Supporting Information Tables S1– www.StemCells.com VC AlphaMed Press 2014 810 Four Signal Pathways in Jaw Blastemal Regeneration

S4). Overall, 5,118 genes were significantly deregulated (q- and 2 dpa, and were involved in epidermis development, value cut-off 0.05) in time between all four time points. angiogenesis, response to stimuli, and protein kinase/phos- Except for 67 genes that were regulated at all stages, wound phatase activity. Until 5 dpa, significantly enriched genes were healing and regeneration temporally altered the expression of restricted to the sarcoglycan complex, and myosin filaments. 2,800 genes between 2 hpa and 0 hpa, 533 genes between 2 Muscle fiber development was enriched at 2 dpa and 5 dpa dpa and 2 hpa, and 240 genes between 5 dpa and 2 dpa (Fig. 3A). (Supporting Information Fig. S4). This decreasing number of deregulated genes nicely paralleled the healing progress of Functional Classification of Highly Enriched GO Term lower jaw regeneration. Subnodes To evaluate the effectiveness of our GO-based annotations In this part of our study, we used DAVID Functional Annota- and differential analyses, we compared our readouts of tion Clustering [18] to select common genes by examining extracellular matrix and chondrogenic signature gene clusters annotations and identifying significantly shared GO terms. We with the data from previous investigations [21, 22]. We found also used the functional classification tool to quickly organize that the expressions of 50 out of 86 genes that are associated large lists of genes into functionally related groups. At 2 hpa, with ECM, chondrogenesis, and skeletal development, and 244 upregulated ribonucleotide binding were accu- 63% of development-associated genes (22/35) were altered mulated. This included 44 small GTP-binding proteins and 75 (Supporting Information Table S5). protein kinases. At 0 hpa, 29 genes that function in intracellu- We then mapped the differentially expressed genes lar protein transport or cellular macromolecule localization (p < .05) to canonical pathways referred from the KEGG [23]. were identified (Supporting Information Table S9). We compared this map with 10 major intracellular signaling When 2 dpa was compared with 2 hpa, 302 genes were pathways previously reported to be key players in initiating upregulated, while 1,705 genes were downregulated (p < .05). and maintaining the regeneration: MAPK, ErbB, Wnt, Notch, We detected a clear difference in upregulated proteins for the Hedgehog, TGF-beta, VEGF, ECM- interaction, cell following GO terms: calcium ion binding, DNA replication, adhesion molecules, and Jak-STAT signaling pathways [24]. All nucleosome assembly, and intermediate filament cytoskeleton. these pathways were successfully identified in our time-lapse The 1,705 downregulated genes were significantly enriched in transcriptomal sequences of lower jaw regeneration. transmembrane, transporter activity, and transcription regula- Previous studies identified 21 genes during intestinal and tor activity. At 5 dpa, 410 genes were upregulated and 50 body wall regeneration of Apostichopus japonicus [24]. In con- were downregulated. The 35 most accumulated genes were trast, our study identified 281 candidate genes from the 1,191 multiple Hox-, Fox-, and Sox-paralogs (Supporting Information genes that are associated with the 10 pathways (Supporting Table S9). Information Table S6), accounting for 23.6% of total signaling GO classification and enrichment not only confirmed the steps. However, statistical analysis revealed that only 4 of the GO terms associated with RNA processing, transcription regu- 10 pathways were significantly enriched (Supporting Informa- lation, GTP binding-protein kinase, transmembrane transport- tion Table S7), suggesting distinct regenerative mechanisms ers, and calcium ion binding, but also functionally assigned responsible for lower jaw regeneration. Together, our tempo- the clustered genes to the superfamily level. We found that ral RNA-sequencing and GO term-annotation successfully iden- 10 superfamilies of Fox, Sox, Pax, POU, Six, Hox, Wnt, Bmp, tified the appropriate number of the deregulated genes, and Fgf, and Collagen enriched 101 deregulated genes (Fig. 3B). thereby strengthened our ground work to further identify key Thus, integration of functional classification into hierarchical signaling cascades responsible for lower jaw regeneration. GO trees permitted us to focus more on the GOs relevant to oxygen metabolism, angiogenesis, transmembrane transport- Genome-Wide Annotation of Differential Transcripts to ers, GTPase-tyrosine kinase activity, DNA replication, neural GO Term Networks crest cell migration, cell differentiation, axon extension, and We annotated the differential transcripts and generated a epidermal and muscle development. tree diagram displaying all possible GO terms in networks. GO terms are organized hierarchically such that higher level terms Prediction of Signaling Cascades Associated with are more general and thus are assigned to more genes, while Regeneration Processes descendant terms are more specific and related to parents. To capture the difference in gene expression patterns and We then identified the accumulation of protein identifiers novel signaling pathways, we applied Euclidean distance mea- assigned to a given GO term, and compared the ratio of pro- surement and performed a similarity search for common teins annotated to a given GO term to the total number of expression patterns of the target genes. Taking advantage of proteins available from the GO database. As an unbiased our identified GO-gene superfamily architecture (Fig. 3), we standard, we limited our GO term search to the top 3% of arbitrarily determined several index genes associated with the highly enriched terms and organized the accumulations important GO terms and regenerative phenotypes, selected according to the regenerative process (Supporting Information the top 30 of the most similarly changed genes (Supporting Table S8). Relative to preamputation (0 hpa), 28 GO terms Information Table S10), and made predictions about the novel were specifically accumulated at 2 hpa. These GO terms were signaling connections. highlighted in acute responses to the damage (Fig. 3A). To characterize the molecular mechanism of cell type con- When comparing 2 dpa with 2 hpa, four temporally versions, we performed cluster analysis of two well-known enriched terms were related to DNA replication: initiation, marker genes associated with pluripotency: pou5f1 and regulation of angiogenesis, heparin binding, and notochord foxo1b. Among the genes differentially expressed between 2 cell differentiation. Eight terms were enriched at both 2 hpa hpa and 0 hpa, the top 30 genes with similar expression

VC AlphaMed Press 2014 STEM CELLS Zhang, Wang, Lyu et al. 811

Figure 3. Integrative analyses of time point-based transcriptome establishes a (GO) network and gene superfamily archi- tecture. (A): Hierarchical GO-analysis reveals accumulation and connection of time point-based GO terms. The upper plate (yellow) shows the genes enriched at all time points. The middle plate shows the specific terms for each time point of the regenerative process. Each time point was indicated by a color (2 hpa in pink, 2 dpa in green, and 5 dpa in blue). The overlapping area shows the enriched genes linking the successive time points. Accumulation of time point-based connecting GO terms parallel to the successive cell type con- versions from re-epitheliation to blastema formation to blastema specification (bottom plate). Panel (B) shows the enriched GO terms and 10 gene superfamilies by integration of hierarchy analysis into David functional classification. These deregulated genes contain inductive signals, blastemal border specifiers, tissue-specific specifiers, and pattern genes. changes in the pou5f1 cluster highly overlapped with the that cdx4, , hox9a/b, hif1, and kdrl were coincidently acti- cdx4. Cdx4-Hoxa9 and Cdx4-Sall4 regulatory modules have vated at the early stage of jaw regeneration (Supporting Infor- been reported to respecify the hematopoietic and hemangio- mation Tables S1 and S2). In addition, activation of hif1,a blast population [25, 26], while gene kdrl, also called flk1,is master regulator of the hypoxia response and angiogenesis expressed in hemangioblasts. kdrl enhancer contains - initiation (Supporting Information Table S10) [28], the pou5f1- binding sites [27]. So, we examined these genes and found cdx4-kdrl regulatory module was proposed to play a role in www.StemCells.com VC AlphaMed Press 2014 812 Four Signal Pathways in Jaw Blastemal Regeneration blastemal angiogenesis and vasculogenesis. foxo1b shared a epidermis (Fig. 4G, 4H), wounding strongly induced Pax3a cluster with pou3f1, foxi1, fhit, and enpp1 at 2 hpa. pou3f1, expression in the basal layer (Figs. 4C, 4D, 5G). also known as Oct-6, is abundantly expressed in the epidermis While Foxi1-expression increased at the surface of the and regulates epidermal cell differentiation [29]. Likewise, wound epidermis and decreased in the presumptive blastema foxi1 is recognized as an epidermal cell specification marker region (3 dpa), Hoxa2b and Pax3a expression increased and [30]. Since enpp1, a member of the ecto-nucleotide pyrophos- spread from the basal layer to the blastema (Fig. 5H, 5I, data phatase/phosphodiesterase family, influences a wide range of not shown). At 7 dpa, Hoxa2b- and Pax3a-expressing mesen- tissues through various signaling pathways [31], it is reasona- chymal blastema encompassed Sox9a-expressing chondrogenic ble to converge foxi1-foxo1b-pou3f1 signaling transduction blastema (Fig. 5J–5M). These observations suggested that epi- with wound epidermis reconstruction. dermis reconstitution and blastema formation can be charac- Consistent with its function of global chromatin modifica- terized by the regional and dynamic expression of Foxi1, tion [32], foxi1 was reactivated and formed a broadly mixed Pax3a, Hoxa2b, and Sox9a/b. signal circuit at 5 dpa, including bhlha15, , tjp2a, and Unlike Foxi1 expression in the surface layer of the epi- myod1 (Supporting Information Table S10). More broadly, the dermis, Mek1/2 was expressed in the epidermal basal layer, foxi1-sox10-myod1 cluster interconnected with the hoxa2b Meckel’s surroundings, and intramuscular connective tissue. and pax3a clusters. This foxi1-hoxa2b-pax3a broad signal cir- Like Foxi1, p-Erk1/2 was expressed in small nucleated cells cuit contained two sox (sox21a), three fox (, ), and and certain fragmented muscle cells in the newly forming seven hox genes. hypodermis between the wound epidermis and muscle (Fig. A myoprogenitor marker, pax3a, closely clustered with 5D–5F). Temporospatial colocalization of Mek1/2 and p-Erk1/ matn3a, which is usually present in the cartilage extracellular 2 with Pax3a and Foxi1 during wound epidermis reconstitu- matrix. We found that coll1a1a, coll1a2, annexin A6, and tgfbi tion and generation of blastema progenitors in the prospec- clustered together. The gene tgfbi encodes an ECM protein, tive mesenchymal blastema suggest that the expression of and is reportedly involved in endochondrial bone formation in Foxi1 and Pax3a not only acts as an active cell identity cartilage muscle fiber growth [33]. Coactivation of coll1a1a, marker, but also regulates cell fate through the MAPK and col112a, col2a1a, and pax3a at 2 hpa and 5 dpa supported ERK pathways. pax3a-mant3a association with cartilage collagens in the wound epidermis and chondrogenic blastema. Cellular Transformation and Tissue Compartments of Isl1 expression marks the multipotent stem cells, giving Three Cell Lineages rise to a broad range of cardiovascular tissues, pharyngeal To visualize blastema formation and cell identity conversions muscle, and the neural crest [34, 35]. Isl1 is also required for in the in vivo environment, we used three transgenic fishes multiple aspects of motor neuron development [36]. At the with fluorescent labeling: flk1:EGFP [37], pou5f1:EGFP [12], initial phase of jaw regeneration, isl1 was upregulated with and isl1:GFP [38]. These lines express GFP under the control wnt11, fgf8b, and cdh7, genes which have kinase and GTPase of the tissue-specific promoter. The GFP expression patterns activity, the ATP-binding cassette, and structural elements of recapitulate the endogenous expression of the corresponding the cytoskeleton, such as riok2, sgk2a, and abcf2. Unexpect- genes (Supporting Information Fig. S5). edly, sox9a clustered with the genes annotating the planar In uncut fish, flk1/kdrl:GFP transgene (hereafter referred cell polarity (PCP) signaling components (fzd7a, prickle1)at5 to as flk1-GFP) was expressed specifically in the blood vessels dpa. It is likely that wnt11 and fgfb8 act as directional cues to in the hypodermis and muscle (Fig. 6A). Angiogenic sprouting regulate the PCP pathway and small GTPase signaling in isl1- of blood vessels occurred in the wound muscle tissues as an positive and sox9a-positive cells during wound epidermis and initial response to amputation at 2 hpa. This response was fol- chondrogenic blastema reorganization. Altogether, we identi- lowed by the emergence of GFP-expressing cells in the newly fied four signal pathways orchestrating lower jaw regenera- regenerating hypodermis (prospective blastema, Fig. 6B, 6C). tion. As shown in Supporting Information Table S10, these Over the following 2 days, GFP-vasculature was transiently pathways are directly or indirectly linked to Rho-GTPase and expressed and GFP-expressing cells increased. These cells con- MAPK pathways. stituted one-third of all blastema cells (Fig. 6D, 6E). At 5 dpa, only a few GFP-positive cells remained in the hypodermis and Wound Epidermis and Blastema Display Characteristic chondrogenic center (Fig. 6F). The sustainable and bright fluo- Expression Patterns rescent signal in blastemal cells and intermittent fluorescence To test our proposed signaling pathways and evaluate the util- in the blastema vasculature was consistent with the previous ity of transcript profiling for prediction of protein expression report that Flk1 expression is stronger in developing angio- levels, we examined the cellular expression patterns of blast/endothelial precursors than in mature vessels [39, 40]. selected several index genes. In the uncut lower jaw, Foxi1 Flk1-GFP positive blastemal cells were not costained by protein was localized primarily in the epidermis and sparsely Foxi1, Sox9a, or Sox9b antibodies (Fig. 6F–6H), presumably in the dermis (Figs. 4A, 5A), whereas Pax3a was mainly dis- representing Flk1-positive hemangioblasts. In pou5f1- tributed in connective tissues (Fig. 4C). Sox9a and Sox9b were promoter-driven GFP transgenic fish, GFP-positive cells were differentially and weakly expressed in the mandible perios- only harbored in the dermis-hypodermis adjacent to the teum and muscle (Fig. 4F, 4G). After amputation, Foxi1 was Meckel’s cartilage (Fig. 4F). After amputation, pou5f1-GFP sig- rapidly induced in wound epidermis and the surrounding nor- nals were detected in the intermuscular vasculature within mal epidermis as well as in the presumptive blastema of small the presumptive blastema region at 14 hpa (Fig. 4H). Thus, nucleated cells and fragmented muscle cells (Figs. 4B, 5B, 5C). the pou5f1 gene was actively promoted in vascular cells. In comparison with the even induction of Sox9b in the wound These observations suggest that wounding induced a double

VC AlphaMed Press 2014 STEM CELLS Zhang, Wang, Lyu et al. 813

Figure 4. Initial induction of five index genes. Compared to preamputation (0 hpa), wounding induced an immediate increase of Foxi1 expression in the wound epidermis and on the surface of the surrounding normal epidermis at 14 hpa (A, B). In isl1-GFP transgenic fish, wounding prompted quick induction of Pax3a in the basal layer of regenerating epidermis (C, D), isl1-GFP in basal layer cells and intramuscular connective tissues like peripheral nerves (C–E), and Pou5f1 in the adjoining area between wound epidermis and wound muscle (E). The inset of (D) highlights isl-GFP positive cells along the basal layer. Panels (F)–(H) show weak expression of Sox9a, Sox9b, and pou5f1-GFP in pou5f1-GFP transgenic fish. Sox9b expression was significantly increased in the wound epidermis (H). Activation of pou5f1-GFP transgene is highlighted in (H) inset. Scale bars (A), (B), (F), (G) 5 100 mm; (C), (D), (E), (H) 5 50 mm. Abbreviation: GFP, green fluorescent protein. process of blastemal angiogenesis and vasculogenesis, Isl1-GFP has been reported to be expressed in the cranial possibly by dedifferentiation of hemangioblast derivatives (Fig. motor and sensory neurons (Supporting Information Fig. S2) 6A–6E). [38]. Here, we found that wounding induced Isl1-driven GFP www.StemCells.com VC AlphaMed Press 2014 814 Four Signal Pathways in Jaw Blastemal Regeneration

Figure 5. Dynamic expression profile of several regeneration-related genes. Foxi1 was expressed in normal epidermis and hypodermis (A). After amputation, Foxi1 expression increased in the surrounding normal epidermis. Wounding also induced Foxi1 and P-Erk1/2 expression in small nucleated cells (sc) and damaged muscles (B, C, E). With an increase on the epidermal surface at 3 dpa, Foxi1 decreased in the blastema region (H). Mek1/2, Hoxa2b, and Pax3a were initially induced in epidermal basal cells (D, G, I), and then increased in the mesenchymal blastema (J, K). During blastema respecification, Sox9a was induced in epidermis and chondrogenic blas- tema (L, M). Panel (F) shows H&E staining of (E). Panels (H) and (I) show antibody-alkaline phosphatase-NBT staining. The remaining fig- ures were stained with fluorescence-conjugate secondary antibodies. Blue indicates DAPI staining. Scale bars (B), (E), (L), (M) 5 25 mm; the rest 5 50 mm. Abbreviations: BL, basal layer of wound epidermis; Coc, chondrogenic center; fmc, fragmented muscle; Mc, Meckel’s cartilage; md, mandible; MsB, mesenchymal blastema; sc, small nucleated cells. expression in the new regenerating basal layer of wound epi- Information Fig. S6). These findings suggest that activation of dermis and blastemal region (Fig. 4C–4E). Initially, a stream of Isl1 signaling may represent dedifferentiation of neuro-neural isl1-GFP expression extended along the basal layer of the crest derivatives and extension of injured nerve fibers. wound regenerating epidermis, partially overlapping with Pax3a (Fig. 4D). At 1 dpa, isl1-GFP largely appeared at Meck- Foxi1 Is Required to Modulate Epidermis el’s cartilage and dotted at the presumptive blastema region Reconstitution and Blastema Formation (Fig. 4E). At 6 dpa, isl1-GFP-expression remained at the chon- We have reported that Foxi1 is required for proper growth of drogenic centers (Coc) (Fig. 6I). We found that isl1-GFP par- the lower jaw during embryonic development and adult tially overlapped with the immunostaining of axonemal a- regeneration [7, 14]. Now, we note that Foxi1 proteins are tubulin. In contrast to more tubulin staining in the interman- mostly distributed in the epidermis, with some in the blas- dibularis posterior area, more isl1-GFP expressing cells were tema. In the Morpholino-control animals, wounding induced observed in the intermandibularis anterior region (Supporting Foxo1b and Pax3a expression, largely in the basement of the

VC AlphaMed Press 2014 STEM CELLS Zhang, Wang, Lyu et al. 815

Figure 6. Visualization of expression of flk1-GFP and isl1-GFP transgenes during blastema regeneration. In flk1-GFP transgenic fish, flk1- expressing cells first occurred in the newly formed hypodermis, and then spread to the vessel-rich muscle; the expression pattern of flk1-GFP mirrored that of 594-daylight-conjugate anti-GFP in immunohistochemistry (A–F). During blastema chondrogenic respecifications (5 dpa, 6 dpa), flk1-GFP, isl1-GFP, Sox9a-, Sox9b-, and Foxi1-expressing cells only remained in the Coc. Obviously, flk1-GFP (green) did not costain with antibodies Sox9b, Sox9a, or Foxi1 (F–I). Scale bars 5 50 mm. Abbreviations: Coc, chondrogenic center; GFP, green fluo- rescent protein. regenerating epidermis at 1 dpa. Foxi1-Morpholinos caused transcription domains. These results suggested that specifica- expansion of Foxo1b- and Pax3a-expressing cells into the tion of sox9a- and hoxa2b-expressing cells to the pharyngeal damaged muscle (Supporting Information Fig. S7A–S7D). Per- arch components requires a downregulation of foxi1. turbation of Foxi1 signaling also impaired blastema vasculo- genesis, without affecting flk1-GFP expression in the surrounding muscle (Supporting Information Fig. S7E, S7F). DISCUSSION These results suggest that Foxi1 not only promotes specifica- Regenerative medicine offers hope to patients by replacing tion of epidermal ectoderm but also inhibits blastema damaged tissues with readily available sources. The most formation. attractive strategy is to convert the available cells into the desired cell types [43]. To fulfill this promise, recent efforts Foxi1, hoxa2b, and sox9a Crossregulate to Reciprocally have focused on the development of biomaterials to aid in Maintain Expression Patterns the healing response and/or to provide a scaffold that can promote appropriate tissue formation [44]. We have shown To trace back to the embryonic compartments of Foxi1- that zebrafish amputation can induce successive cellular trans- expressing cells, we examined the embryonic expression pat- formation and create two transitional blastemal structures/ terns of foxi1, hoxa2b, and sox9a/b by combining RNA knock- mesenchymal and chondrogenic blastema. In this study, we down techniques with whole mount RNA in situ hybridization could not clearly identify which biomaterials would trigger (Fig. 7). In agreement with the previous reports, we found successive cell-based regeneration after lower jaw injury. that all four genes are expressed on the midbrain and/or However, our integrative RNA-sequencing analyses identified hindbrain and pharyngeal arches during embryonic jaw devel- four novel signaling pathways which orchestrate the progres- opment (48–55 hpf) [14, 41, 42]. Microinjection of antisense sive cell-type conversions and tissue reorganization. We pro- sox9a-Mo caused a reduction of both foxi1 (Fig. 7A, 7B) and pose the following model for blastema regeneration of the hoxa2b domains (Fig. 7D, 7E) in the pharyngeal arches, while lower jaw. their expression in the mid-hind brain was not altered. hoxa2b morphants displayed an expansion of sox9a domain in myelencephalon (Fig. 7G, 7H) and a slight diffusion of foxi1(- 1. Foxi1 and Pax3a signaling axes coordinately orchestrate wound epidermis reconstruction and blastema formation. Fig. 7A, 7C). In the foxi1-Mo embryos, however, the express- During lower jaw regeneration, an initial response (0–24 ing domains of both hoxa2b (Fig. 7D, 7F) and sox9a (Fig. 7G, hpa) to wound signaling is collagen deposition, correspond- 7I) were strikingly expanded in the branchial arches, and ing with upregulation of col11a1a, col11a2, and col2a1a showed a Mo-foxi1 concentration-dependent effect (Support- genes. These activities include Foxi1-expression on the sur- ing Information Fig. S8). In accordance with Foxi1 protein’s face layer of the epidermis, Pax3a- and Hoxa2b-expression inhibitory effects on Foxo1b1 and Pax3a1 cell expansion, in the basal layer of epidermis, and Sox9b-expression on foxi1 transcript represses expansion of hoxa2b- and sox9a- the whole epidermis. Various types of cells accumulate www.StemCells.com VC AlphaMed Press 2014 816 Four Signal Pathways in Jaw Blastemal Regeneration

Figure 7. Analyses of foxi1, sox9a, and hoxa2b expression by whole mount embryo in situ hybridization combined with morpholino injection. The standard control morpholino (A,D,G), or the gene-specific morpholino of either foxi1-Mo (F, I), hoxa2b-Mo (C, H),or sox9a-Mo (B, E) was separately microinjected into embryos 48–55 hpf (hour postfertilization). Compared with microinjection of standard control morpholino, the changes of expression patterns are shown in (A–C) for foxi1; in (D–F) for hoxa2b; and (G-I) for sox9a. Although sox9b is also expressed in mid-hind brain and pharyngeal arches, its expression is scarcely altered by foxi1-Mo, hoxa2b-Mo, or sox9a- Mo (data not shown). Each microinjection contains 1 ml of 0.1 mM morpholino. Scale bars 5 250 mm. along the deposited collagen matrix. The regional and and cells derived from the neural crest [50, 51]. Our cell dynamic expression of Foxi1, Pax3a, Hoxa2b, Sox9a/b, morphology and differentiation analyses have indicated that Pou5f1, and Isl1 indicates that Pax3a1 basal layer cells multiple cell sources, including blood-derivative cells, are arise from the interactions between immigrated epidermal dedifferentiated into blastemal progenitors [7]. In this study, cells and the wound mesodermal tissues, and subsequently we supplemented blastema formation with Pax3a-mediated differentiate into Foxi11/Sox9b1 epidermal cells or cell type conversions. Additionally, pluripotent-associated Hoxa2b1/Pax3a1 blastemal cells. The multipotent charac- factor pou5f1 grouped with hematopoietic activator cdx4 teristic of regenerating basal layer cells resembles the and endothelium-specific receptor tyrosine kinase kdrl gene. regenerative capability of EPI-NCSC [45]. This pathway is supposed to regulate hemangioblast-derived How do Pax3a-expressing mesenchymal cells became multi- blastemal angiogenesis and vasculogenesis. potent epidermal cells? In mice, activation promotes 3. Neuronal PCP signaling pathways regulate positional pat- the differentiation of mesenchymal stem cells toward the terning in blastema regeneration. myogenic lineage and regulates mesenchymal-epithelial It has been reported that the positional identity of the blas- interconversion [46–48]. In zebrafish mandible regeneration, tema correlates with the characteristic expression pattern of we propose that the pax3a-mant3a-cartilage collagen path- Hox genes [7] and FoxD [52]. Now, we find that wnt11-PCP way designs cell-type conversions. Different from skin devel- pathways are induced during re-epitheliation and chondro- opment in which col1a2 transcript is strongly expressed in genic blastema respecifications. In the initial phase, the wnt11- the basal layer of epidermal and dermal endothelial cells PCP pathway components direct asymmetric distribution of [49], expression of cartilaginous collagen type XI and type II isl1 signaling in the injured cranial nerves and muscles (Sup- in regenerating tissues can provide structural support, and porting Information Fig. S5). During blastema respecification (5 orientation for epidermal cells and blastemal cells. dpa), the wnt11r-PCP pathway links to sox9a in blastema tran- A related question is why blastema formation proceeds from sient chondrogenesis. Given that core PCP genes are crucial re-epitheliation. Zebrafish epidermis contains diverse cell for the development and function of the nervous system and types, including a variety of subtypes of ionocytes. Foxi1 reg- are involved in neural tube closure, ependymal polarity, neuro- ulates H1-ATPase-rich cell differentiation [20]. From thou- nal migration, and axon guidance [53, 54], the Isl1-wnt11-PCP- sands of deregulated genes, we identified foxi1-foxo1b- sox9a pathways could provide instructions for the cartilage pou3f1 cluster signals. This pathway may promote differentia- matrix in tissue patterning and polarize the motile behaviors tion of Pax3a1 progenitors into Foxi1-expressing epidermal of blastemal cells with respect to the anteroposterior axis. cells. In the blastema, Foxi1 represses expansion of hoxa2b/ pax3a, and favors mesenchymal-chondrogenic blastema tran- sition. This signal switch can explain how Foxi1 alternatively CONCLUSIONS promotes re-epitheliation before blastema respecification. 2. pou5f1-cdx4-kdrl signaling pathway regulates blastemal According to the dynamic signaling changes and successive angiogenesis and vasculogenesis. cellular transformation during regeneration of zebrafish lower Most published studies suggest that blastema formation fol- jaws, we propose a jaw-regeneration gene network that con- lows a process of dedifferentiation of mesodermal tissues verges the pouf51-cdx4-kdrl, pax3a-mant3a-col11, isl1-wnt11-

VC AlphaMed Press 2014 STEM CELLS Zhang, Wang, Lyu et al. 817

PCP-sox9a, foxi1-foxo1b-pou3f1 signaling axes onto small tion Initiative Program of Shanghai Ocean University GTPase-MAPK pathways to define wound epidermis recon- (A2302110002) to J.Y, and Shanghai Universities First-class Dis- struction, blastema formation, and blastemal cell lineage spec- ciplines Project of Fisheries. ification. Further studies of these pathways will identify the crucial molecular drivers involved and potential therapeutic targets. This study therefore provides a signaling roadmap to AUTHOR CONTRIBUTIONS study vascular tissue engineering and skin stem cell-based J.Y.: conception and design, financial support, data analysis regenerative therapy. and interpretation, and manuscript writing; H.Z., X.W., K.L., and S.G.: collection and/or assembly of data and data analysis ACKNOWLEDGMENTS and interpretation; C.F.: provision of study material; G.W.: RNA-seq data analysis; X.Z.: manuscript writing. H.Z., X.W., We thank Kathy Kyler for editing the manuscript. We appreci- and K.L. are joint first authors. ate Wei Zhen, Ying Zhang, Qin Yang, Junhui Cao, and Xiaoyun Zhou for their technical assistance. We are indebted to Guan- jie Chen and Suiyuan Zhang for their valuable computational analysis in RNA deep sequencing. This work was supported in DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST part by the Innovation Program of Shanghai Municipal Educa- The authors indicate no potential conflicts of interest. tion Commission (13ZZ126) to J.Y., the International Coopera-

REFERENCES ovary-testis transformation. Cell Death Dis chem Physiol Part D Genomics Proteomics 2013;4:e930. 2011;6:195–205. 1 Poss KD. Advances in understanding tis- 14 Nissen RM, Yan J, Amsterdam A et al. 25 Davidson AJ, Ernst P, Wang Y et al. cdx4 sue regenerative capacity and mechanisms in Zebrafish foxi one modulates cellular mutants fail to specify blood progenitors and animals. Nat Rev Genet 2010;11:710–722. responses to Fgf signaling required for the can be rescued by multiple hox genes. 2 Whited JL, Tabin CJ. Regeneration review integrity of ear and jaw patterning. Develop- Nature 2003;425:300–306. reprise. J Biol 2010;9:15. ment 2003;130:2543–2554. 26 Yan J, Chen YX, Desmond A et al. Cdx4 3 Fisher MB, Mauck RL. Tissue engineering 15 Trapnell C, Hendrickson DG, Sauvageau and menin co-regulate Hoxa9 expression in and regenerative medicine: Recent innova- M et al. Differential analysis of gene regula- hematopoietic cells. PLoS One 2006;1:e47. tions and the transition to translation. Tissue tion at transcript resolution with RNA-seq. 27 Ishitobi H, Wakamatsu A, Liu F et al. Eng Part B Rev 2013;19:1–13. Nat Biotechnol 2013;31:46–53. Molecular basis for Flk1 expression in 4 Nemeno-Guanzon JG, Lee S, Berg JR hemato-cardiovascular progenitors in the et al. Trends in tissue engineering for blood 16 Young MD, Wakefield MJ, Smyth GK et al. Gene ontology analysis for RNA-seq: mouse. Development 2011;138:5357–5368. vessels. J Biomed Biotechnol 2012;2012: 28 Dewhirst MW, Cao Y, Li CY et al. Explor- 956345. Accounting for selection bias. Genome Biol 2010;11:R14. ing the role of HIF-1 in early angiogenesis 5 Nowak JA, Polak L, Pasolli HA et al. Hair and response to radiotherapy. Radiother follicle stem cells are specified and function 17 Yekutieli DBY. Resampling-based false discovery rate controlling multiple test proce- Oncol 2007;83:249–255. in early skin morphogenesis. Cell Stem Cell 29 Faus I, Hsu HJ, Fuchs E. Oct-6: A regula- 2008;3:33–43. dures for correlated test statistics. J Stat Plan Inference 1999;82 171–196. tor of keratinocyte gene expression in strati- 6 Tasoglu S, Demirci U. Bioprinting for fied squamous epithelia. Mol Cell Biol 1994; 18 Huang da W, Sherman BT, Lempicki RA. stem cell research. Trends Biotechnol 2013; 14:3263–3275. Systematic and integrative analysis of large 31:10–19. 30 Kwon HJ, Bhat N, Sweet EM et al. Iden- 7 Wang X, He H, Tang W et al. Two origins gene lists using DAVID bioinformatics resour- ces. Nat Protoc 2009;4:44–57. tification of early requirements for preplaco- of blastemal progenitors define blastemal dal ectoderm and sensory organ regeneration of zebrafish lower jaw. PLoS 19 Huang da W, Sherman BT, Zheng X et al. development. PLoS Genet 2010;6:e1001133. Extracting biological meaning from large One 2012;7:e45380. 31 Mackenzie NC, Huesa C, Rutsch F et al. gene lists with DAVID. Curr Protoc Bioinfor- 8 Yannas IV. Emerging rules for inducing New insights into NPP1 function: Lessons matics 2009;Chapter 13:Unit 13 11. organ regeneration. Biomaterials 2013;34: from clinical and animal studies. Bone 2012; 321–330. 20 Chang WJ, Hwang PP. Development of 51:961–968. 9 Borchardt T, Looso M, Bruckskotten M zebrafish epidermis. Birth Defects Res C 32 Yan J, Xu L, Crawford G et al. The fork- et al. Analysis of newly established EST data- Embryo Today 2011;93:205–214. head FoxI1 remains bases reveals similarities between heart 21 Andreasen EA, Mathew LK, Tanguay RL. bound to condensed mitotic regeneration in newt and fish. BMC Regenerative growth is impacted by TCDD: and stably remodels chromatin structure. Genomics 2010;11:4. Gene expression analysis reveals extracellular Mol Cell Biol 2006;26:155–168. 10 Monaghan JR, Epp LG, Putta S et al. matrix modulation. Toxicol Sci 2006;92:254– 33 Kim HR, Ingham PW. The extracellular Microarray and cDNA sequence analysis of 269. matrix protein TGFBI promotes myofibril bun- transcription during nerve-dependent limb 22 Herlofsen SR, Kuchler AM, Melvik JE dling and muscle fibre growth in the zebra- regeneration. BMC Biol 2009;7:1. et al. Chondrogenic differentiation of human fish embryo. Dev Dyn 2009;238:56–65. 11 Qin YF, Fang HM, Tian QN et al. Tran- bone marrow-derived mesenchymal stem 34 Bu L, Jiang X, Martin-Puig S et al. scriptome profiling and digital gene expres- cells in self-gelling alginate discs reveals Human ISL1 heart progenitors generate sion by deep-sequencing in normal/ novel chondrogenic signature gene clusters. diverse multipotent cardiovascular cell line- regenerative tissues of planarian Dugesia Tissue Eng Part A 2011;17:1003–1013. ages. Nature 2009;460:113–117. japonica. Genomics 2011;97:364–371. 23 Kanehisa M, Goto S, Kawashima S et al. 35 Harel I, Nathan E, Tirosh-Finkel L et al. 12 Parvin MS, Okuyama N, Inoue F et al. The KEGG resource for deciphering the Distinct origins and genetic programs of Autoregulatory loop and retinoic acid repres- genome. Nucleic Acids Res 2004;32:D277– head muscle satellite cells. Dev Cell 2009;16: sion regulate pou2/pou5f1 gene expression 280. 822–832. in the zebrafish embryonic brain. Dev Dyn 24 Sun L, Chen M, Yang H et al. Large scale 36 Liang X, Song MR, Xu Z et al. Isl1 is 2008;237:1373–1388. gene expression profiling during intestine required for multiple aspects of motor neu- 13 Sun D, Zhang Y, Wang C et al. Sox9- and body wall regeneration in the sea ron development. Mol Cell Neurosci 2011;47: related signaling controls zebrafish juvenile cucumber Apostichopus japonicus. Comp Bio- 215–222. www.StemCells.com VC AlphaMed Press 2014 818 Four Signal Pathways in Jaw Blastemal Regeneration

37 Choi J, Dong L, Ahn J et al. FoxH1 nega- and pectoral fin development. Development 49 Le Guellec D, Morvan-Dubois G, Sire JY. tively modulates flk1 gene expression and 2005;132:1069–1083. Skin development in bony fish with particular vascular formation in zebrafish. Dev Biol 43 Cohen DE, Melton D. Turning straw into emphasis on collagen deposition in the der- 2007;304:735–744. gold: Directing cell fate for regenerative med- mis of the zebrafish (Danio rerio). Int J Dev 38 Higashijima S, Hotta Y, Okamoto H. Visu- icine. Nat Rev Genet 2011;12:243–252. Biol 2004;48:217–231. alization of cranial motor neurons in live 44 Fisher MB, Mauck RL. Starting with 50 Nakatani Y, Kawakami A, Kudo A. Cellu- transgenic zebrafish expressing green fluores- form, emerging with function: Nanofibrous lar and molecular processes of regeneration, cent protein under the control of the islet-1 scaffolds for tissue engineering of orthopedic with special emphasis on fish fins. Dev promoter/enhancer. J Neurosci 2000;20:206– tissues. Nanomedicine (Lond) 2013;8:505– Growth Differ 2007;49:145–154. 218. 508. 51 Satoh A, Graham GM, Bryant SV et al. Neurotrophic regulation of epidermal dedif- 39 Jin SW, Beis D, Mitchell T et al. Cellular 45 Sieber-Blum M, Hu Y. Epidermal neural ferentiation during wound healing and limb and molecular analyses of vascular tube and crest stem cells (EPI-NCSC) and pluripotency. regeneration in the axolotl (Ambystoma mex- lumen formation in zebrafish. Development Stem Cell Rev 2008;4:256–260. 2005;132:5199–5209. icanum). Dev Biol 2008;319:321–335. 46 Gang EJ, Bosnakovski D, Simsek T et al. 52 Scimone ML, Lapan SW, Reddien PW. A 40 Liao W, Bisgrove BW, Sawyer H et al. Pax3 activation promotes the differentiation forkhead transcription factor is wound- The zebrafish gene cloche acts upstream of a of mesenchymal stem cells toward the myo- induced at the planarian midline and flk-1 homologue to regulate endothelial cell genic lineage. Exp Cell Res 2008;314:1721– required for anterior pole regeneration. PLoS differentiation. Development 1997;124:381– 1733. Genet 2014;10:e1003999. 389. 47 Wiggan O, Fadel MP, Hamel PA. Pax3 53 Tatin F, Taddei A, Weston A et al. Planar 41 Hunter MP, Prince VE. Zebrafish hox induces cell aggregation and regulates phe- cell polarity protein Celsr1 regulates endo- paralogue group 2 genes function redun- notypic mesenchymal-epithelial interconver- thelial adherens junctions and directed cell dantly as selector genes to pattern the sec- sion. J Cell Sci 2002;115:517–529. rearrangements during valve morphogenesis. ond pharyngeal arch. Dev Biol 2002;247:367– 48 Wiggan O, Shaw AE, Bamburg JR. Essen- Dev Cell 2013;26:31–44. 389. tial requirement for Rho family GTPase sig- 54 Tissir F, Goffinet AM. Shaping the nerv- 42 Yan YL, Willoughby J, Liu D et al. A pair naling in Pax3 induced mesenchymal- ous system: Role of the core planar cell of Sox: Distinct and overlapping functions of epithelial transition. Cell Signal 2006;18: polarity genes. Nat Rev Neurosci 2013;14: zebrafish co-orthologs in craniofacial 1501–1514. 525–535.

See www.StemCells.com for supporting information available online.

VC AlphaMed Press 2014 STEM CELLS