Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jul 3.
Published in final edited form as: J Exp Zool B Mol Dev Evol. 2009 Jun 15;312B(4):343–350. doi: 10.1002/jez.b.21274

Epithelial-Specific Requirement of FGFR2 Signaling During Tooth and Palate Development

RYOICHI HOSOKAWA 1, XUEMEI DENG 1, KAZUNORI TAKAMORI 2, XUN XU 1, MARK URATA 1, PABLO BRINGAS 1, YANG CHAI 1,*
PMCID: PMC2896559  NIHMSID: NIHMS214223  PMID: 19235875

Abstract

Reciprocal interactions between epithelium and mesenchyme are crucial for embryonic development. Fibroblast growth factors (FGFs) are a growth factor family that play an important role in epithelial–mesenchymal tissue interaction. We have generated epithelial-specific conditional knockout mice targeting Fibroblast growth factor receptor 2 (Fgfr2) to investigate the function of FGF signaling during craniofacial development. K14-Cre;Fgfr2fl/fl mice have skin defects, retarded tooth formation, and cleft palate. During the formation of the tooth primordium and palatal processes, cell proliferation in the epithelial cells of K14-Cre;Fgfr2fl/fl mice is reduced. Thus, FGF signaling via FGFR2 in the epithelium is crucial for cell proliferation activity during tooth and palate development.


Fibroblast growth factor (FGF) is one of the growth factors that are involved in intercellular signaling during embryogenesis (Thesleff and Sharpe, ’97; Hogan, ’99). To date, 22 FGF ligands and four FGF receptors have been identified (Ornitz and Itoh, 2001; Itoh and Ornitz, 2004). FGFR1, 2, and 3 contain three extracellular, immunoglobulin-like (Ig-like) domains, an acid box (between Ig-like I and II), a single transmembrane domain, and paired intracellular tyrosine kinase domains. Alternative mRNA splicing of the Fgfr gene specifies the sequence of the carboxyl-terminal half of Ig-domain III, called either IIIb or IIIc (Miki et al., ’92; Chellaiah et al., ’94; Naski and Ornitz, ’98). Exon IIIb is expressed in epithelial lineages, whereas exon IIIc tends to be expressed in mesenchymal lineages (Avivi et al., ’93; Scotet and Houssaint, ’95).

FGF signaling is involved in craniofacial development such as that of the craniofacial bone, palate, salivary gland, tooth, and craniofacial muscle (Nie et al., 2006). During tooth development, Fgfr2b (IIIb) is mainly expressed in the dental and palatal epithelia and Fgfr2c (IIIC) is expressed in the dental mesenchyme (Kettunen et al., ’98). FGF10, which is the ligand for FGFR2b, is expressed in the dental and palatal mesenchyme (Harada et al., 2002; Rice et al., 2004). Moreover, the phenotypes of Fgf10 conventional knockout mice are consistent with those of Fgfr2b mice including retardation of limbs, lungs, salivary glands, and thymus (Sekine et al., ’99; De Moerlooze et al., 2000; Ohuchi et al., 2000). These observations imply that reciprocal interactions between mesenchymal and epithelial tissue play a role in this process.

There are exceptions to these patterns of receptor expression that occur in tooth development. For instance, Fgfr2b is expressed in both ameloblasts and odontoblasts at the newborn stage, and Fgfr2c is expressed in the inner dental epithelium at the bell stage (E16-17) (Kettunen et al., ’98). Furthermore, in the absence of Fgfr2b, Fgf10 can bind to other isoforms of the FGF receptor, such as FGFR1b (Beer et al., 2000). To examine these discrepancies, we created mice lacking all isoforms of Fgfr2 specifically in the epithelium (K14-Cre;Fgfr2fl/fl). K14-Cre;Fgfr2fl/fl mice have defects in skin, teeth, plate, and hindlimbs, but not forelimbs. During tooth and palatal development, epithelial-specific loss of Fgfr2 causes defects in cell proliferation in epithelial of tooth and palate. Organization of the rugae of the palate, which is considered a landmark of palatal development (Cui et al., 2005), is also compromised in K14-Cre;Fgfr2fl/fl mouse.

MATERIALS AND METHODS

Generation of K14-Cre;Fgfr2fl/fl mutant mice

All animal studies were performed according to IACUC guidelines. K14-Cre transgenic mice have been described previously (Indra et al., 2000; Andl et al., 2004). We crossed K14-Cre;Fgfr2fl/+ with Fgfr2fl/fl mice to generate K14-Cre;Fgfr2fl/fl null alleles that were genotyped using PCR primers as previously described (Yu et al., 2003).

Two-component genetic system for marking the progeny of epithelial cells

The R26R conditional reporter allele has been described previously (Soriano, ’99). We mated K14-Cre and R26R mice to generate K14-Cre;R26R embryos. Detection of β-galactosidase activity in whole embryos (E10.5) was carried out as previously described (Chai et al., 2000).

Staining of whole skeleton

Whole skeletal preparations of newborn mice were prepared and stained with Alizarin Red and Alcian Blue as previously described (McLeod, ’80).

Histological analysis

Tissues were fixed in 4% paraformaldehyde in phosphate buffered saline, decalcified in Decalcifying solution (Richard-Allan Scientific, Kalamazoo, MI), paraffin embedded, sectioned at 5 mm, and stained with hematoxylin/eosin.

BrdU labeling/histology

A 10 mg/mL stock of bromodeoxyuridine (BrdU; Sigma, St Louis, MO) was injected intraperitoneally into mice at E13.5 at a dose of 100 μg BrdU per gram of body weight. Mice were sacrificed 1 hr after injection and bones were harvested. BrdU staining was carried out on paraffin sections by using a BrdU staining kit according to the manufacturer's directions (Zymed, South San Francisco, CA).

In situ hybridization

Whole-mount and sectioned in situ hybridizations were performed according to standard procedure (Wilkinson, ’98). Several negative controls (sense probe and no probe) were run in parallel with the experimental reaction. RNA probes were generated as reported previously: Fgfr2b (Sasaki et al., 2006) and Shh (Hui and Joyner, ’93).

RESULTS

K14-Cre;Fgfr2fl/fl mice have skin and skeletal defects

We generated an epithelial-specific gene deletion of Fgfr2 in mice using Cre recombination driven by the epithelial-specific K14 promoter (Andl et al., 2004). K14-Cre;Fgfr2fl/1 mice were indistinguishable from their wild-type littermates (Fig. 1A and C). We crossed K14-Cre;Fgfr2fl/+ mice with Fgfr2fl/fl mice and recovered approximately 25% K14-Cre;Fgfr2fl/fl mice, implying that no significant lethality occurred before birth. K14-Cre;Fgfr2fl/fl mice died within 24 hr of birth without feeding, likely owing to their cleft palate (Fig. 2). The eyes of K14-Cre;Fgfr2fl/fl mice were open owing to the absence of eyelids, and they had shiny skin (Fig. 1B). We detected limb dysgenesis in the hindlimb but not in the forelimb (Fig. 1A–D). Bone staining of K14-Cre;Fgfr2fl/fl mice revealed no bones in the lower limb except the pelvic girdle (Fig. 1E and F).

Fig. 1.

Fig. 1

K14-Cre;Fgfr2fl/fl mice show skin and skeletal defects. (A–D) Macroscopic appearance of newborn control (A, C; K14-Cre;Fgfr2fl/+) and K14-Cre;Fgfr2fl/fl (B, D) mice. Note the shiny skin, open eye, and retarded hindlimb of K14-Cre;Fgfr2fl/fl mice. (E, F) Alizarin red/Alcian blue-stained control (E; K14-Cre;Fgfr2fl/+) and K14-Cre;Fgfr2fl/fl (F) skeletons. K14-Cre;Fgfr2fl/fl mice possess the pelvic girdle but not limb skeleton. (G, H) In situ hybridization of Fgfr2b expression (arrow) in hair follicle of control (G; K14-Cre;Fgfr2fl/+). There is no detectable Fgfr2 expression in K14-Cre;Fgfr2fl/fl mice (H) at E12.5. (I, J) Whole mount X-gal staining of K14-Cre;R26R mice at E10.5 (I) and E11.5 (J). (I) The hindlimb, but not the forelimb, of K14-Cre;R26R mice exhibit strong X-gal staining at E10.5. (J) Oral epithelium exhibit strong X-gal staining (arrow). (K) Section X-gal staining at E12.5. X-gal staining is detectable in dental (arrowhead) and palatal (arrow) epithelium. Scale bars 5 mm in A–F, 200 μm in G, H, K and 1 mm in I.

Fig. 2.

Fig. 2

Tooth development is retarded in K14-Cre;Fgfr2fl/fl mice. (A–F) Hematoxylin and eosin staining at 12.5 (A, B), at 13.5 (C, D), and at 14.5 (E, F). (G, H) Whole mount in situ of Fgf8 at E11.5 maxillary process. (I, J) BrdU staining of mandibula primordium at E13.5. (A, C, E, G, I) K14-Cre;Fgfr2fl/+ (control) mice, (B,D,F,H,J) K14-Cre;Fgfr2fl/fl mice. (A, B) The dental lamina formation of K14-Cre;Fgfr2fl/fl mice is indistinguishable for control mice. Dotted lines indicate the basal lamina. (C, D) The dental epithelium of control mice is bud stage but is still lamina stage in K14-Cre;Fgfr2fl/fl mice (D, arrow). (E, F) Control mice show the cap stage of epithelium but K14-Cre;Fgfr2fl/fl mice indicate the retardation of dental epithelium (F, arrow). (G) The presumptive dental epithelium express Fgf8 in K14-Cre;Fgfr2fl/+ mice (black arrow). (H) Fgf8 expression was reduced in the process of K14-Cre;Fgfr2fl/fl mice (black arrow). (I, J) Dental epithelium exhibit the BrdU positive cells in K14-Cre;Fgfr2fl/+ mice but not in K14-Cre;Fgfr2fl/fl mice. Dotted line indicates the basal lamina. (K) BrdU labeling index of E13.5 sections from K14-Cre; Fgfr2flox/+ (Control) and K14-Cre; Fgfr2flox/flox (conditional knock out; C.K.O.) mice. Five randomly selected, nonoverlapping samples were used from each experimental group. Student t-tests were used for statistical analysis. *: P<0.05. (N = 5). Scale bars 100 μm in A–F, I, J, and 2 mm in (G, H).

To confirm the tissue-specific deletion of Fgfr2 gene expression, we performed in situ hybridization. The epithelium and primordium of the hair follicle expressed Fgfr2b at E12.5 (Fig. 1G, arrow). In contrast, Fgfr2b expression was not detectable in K14-Cre;Fgfr2fl/fl mice (Fig. 1H), consistent with successful epithelial-specific loss of Fgfr2b. Interestingly, K14-Cre;Fgfr2fl/fl mice had limb defects only in the hindlimb, although Fgfr2b and Fgf10 conventional knockout mice form neither fore- nor hindlimbs. To confirm the timing of Cre recombination under the control of the K14 promoter, we crossed K14-Cre mice with Rosa26R mice (Soriano, ’99) and performed X-gal staining of the embryo. E10.5 samples clearly indicated that Cre recombination occurred earlier in the hindlimb than in the forelimb (Fig. 1I). At later stages, Cre recombination was detectable in all epithelial cells (Fig. 1J). This time gap might be a critical reason that the forelimb escaped having defects in formation in K14-Cre;Fgfr2fl/fl mice. We also confirmed the expression pattern in the oral epithelium of the K14 promoter of our construct. K14-Cre;R26R mice exhibited mosaic X-gal staining of the first branchial arch at E10.5 (Fig. 1I, arrowhead) (Chai and Maxson, 2006). At E11.5, we detected strong X-gal staining in the oral epithelium (Fig. 1J). At E12.5, the dental and palatal epithelium are strongly positive for X-gal staining (Fig. 1K) (Xu et al., 2006, 2008).

Tooth formation is compromised in K14-Cre; Fgfr2fl/fl mice owing to reduced cell proliferation

Newborn K14-Cre;Fgfr2fl/fl mice lacked teeth (data not shown). At E12.5, the lamina stage, we detected thickened epithelium in the presumptive tooth development area of K14-Cre;Fgfr2fl/fl mice, which was indistinguishable from that of control mice (Fig. 2A and B). At E13.5, the teeth of control mice had reached the bud stage, but the dental epithelium of K14-Cre;Fgfr2fl/fl mice still appeared to be in the lamina stage (Fig. 2C and D). At E14.5, control mice had reached the cap stage (Fig. 2E), but tooth development remained retarded in K14-Cre;Fgfr2fl/fl mice (Fig. 2F).

To investigate possible mechanisms of this tooth retardation, we analyzed gene expression of candidates expressed in the dental epithelium. Fgf8 is one of the molecules indicating the onset of tooth development (Zhang et al., 2003). Fgf8 was expressed in the presumptive tooth region in control mice at E11.5, whereas it was dramatically reduced in K14-Cre;Fgfr2fl/fl mice, even though Fgf8 expression in the nasal pit area was indistinguishable in K14-Cre;Fgfr2fl/fl and control mice (Fig. 2G and H, open arrowheads). Pitx2, another marker for tooth initiation (Chai and Maxson, 2006), was unchanged in K14-Cre;Fgfr2fl/fl mice relative to control (data not shown).

Next, we analyzed cell proliferation activity in the lamina and bud stages. At E12.5 (lamina stage), cell proliferation activity of K14-Cre;Fgfr2fl/fl mice was indistinguishable from that of control mice in both epithelial and mesenchymal cells (data not shown). At E13.5 (bud stage), we detected a reduction of cell proliferation activity in dental epithelial cells of K14-Cre;Fgfr2fl/fl mice compared with those of control mice, although there was no change in proliferation in dental mesenchymal cells (Fig. 2I–K).

K14-Cre; Fgfr2fl/fl mice develop cleft palate with compromised rugae formation

We observed complete cleft palate in 75% of K14-Cre;Fgfr2fl/fl mice (Fig. 3B), and the remaining 25% displayed submucous cleft palate at the newborn stage. We were unable to detect rugae formation in K14-Cre;Fgfr2fl/fl mice, although rugae were clearly visible on the hard palate of newborn mice (Fig. 3A, black arrows, and 3B). To confirm the presence or absence of rugae, we examined Shh expression, which is a marker for rugae formation (Sasaki et al., 2007), using whole mount in situ hybridization. We detected seven rugae on the secondary palate of control mice at E15.5, based on Shh expression (Fig. 3C). In contrast, Shh expression appeared dotted on the secondary palate of K14-Cre;Fgfr2fl/fl mice, implying that the rugae were not well organized (Fig. 3D). Interestingly, hair follicle formation in the nose in K14-Cre;Fgfr2fl/fl mice was well organized when compared with control mice (Fig. 3C and D). Histological analysis demonstrated that the palatal shelves of K14-Cre;Fgfr2fl/fl mice transformed from vertical to horizontal positions, but the left and right sides of the palatal shelves did not contact each other (Fig. 3F). We hypothesized that cleft palate was owing to insufficient palatal shelf growth, which might be the result of a reduction of mesenchymal cell proliferation. We did detect a reduction in cell proliferation in K14-Cre;Fgfr2fl/fl palatal epithelium at E13.5. We conclude that the reduction of epithelial cell caused the cleft palate in K14-Cre;Fgfr2fl/fl mice.

Fig. 3.

Fig. 3

The cleft palate of K14-Cre;Fgfr2fl/fl mice. (A, B) Macroscopic appearance of hard palate at newborn stage. (C, D) Whole mount in situ of Shh of the secondary palate at E15.5. (E, F) Hematoxylin and eosin staining at E15.5. (G, H) BrdU staining of palatal shelf at E13.5. (A) K14-Cre;Fgfr2fl/+ mice exhibit the rugae formation on the secondary palate (black arrows). (B) K14-Cre;Fgfr2fl/fl mice display the cleft palate without rugae formation (white arrows). (C) K14-Cre;Fgfr2fl/+ mice exhibit well organized Shh expression consisted with the rugae formation (Black arrows). (D) Shh is compromised in K14-Cre;Fgfr2fl/fl mice (white arrows). (E, F) K14-Cre;Fgfr2fl/fl mice exhibit the cleft palate although palatal shelf is horizontal position. (G) The palatal epithelium of K14-Cre;Fgfr2fl/+ mice exhibit BrdU positive cells (black arrows). (H, I) The BrdU positive cells is reduced in K14-Cre;Fgfr2fl/fl mice (white arrows). (I) BrdU labeling index of E13.5 sections from K14-Cre; Fgfr2flox/+ (Control) and K14-Cre; Fgfr2flox/flox (conditional knock out; C.K.O.) mice. Five randomly selected, nonoverlapping samples were used from each experimental group. Student t-tests were used for statistical analysis. *: P<0.05. (N = 5). Scale bars 2 mm in A–D, I, J, and 400 μm in E, F, and 50 μm in G, H.

Furthermore, we investigated whether morphological changes of the palatal shelf occurred before shelf elevation owing to the reduction of epithelial cell proliferation. At E13.5, the size of the palatal shelves of K14-Cre;Fgfr2fl/fl mice was reduced from anterior to posterior when compared with that of control mice (Fig. 4A–D). The shape of the palatal shelves in K14-Cre;Fgfr2fl/fl mice was also compromised. The palatal shelves of K14-Cre;Fgfr2fl/+ (control) mice formed an asymmetric, fingerlike shape (Fig. 4A and C), whereas this asymmetry was largely lost in K14-Cre;Fgfr2fl/fl mice (Fig. 4B and D).

Fig. 4.

Fig. 4

Morphological changes of the palatal shelves before elevation in K14-Cre;Fgfr2fl/fl mice. (A–D) Hematoxylin and eosin staining in K14-Cre;Fgfr2fl/+ (control; A, C) and K14-Cre;Fgfr2fl/fl (B, D) mice at E13.5. A and B are sections from the anterior and C and D are sections from the posterior regions of the palatal shelves. Scale bars 200 μm in A–D.

DISCUSSION

In this study, we examined the biological significance of FGF signaling through Fgfr2 in epithelial cells using a tissue-specific deletion of the Fgfr2 gene. Through the analysis of conditional knockout (K14-Cre;Fgfr2fl/fl) mice, we conclude that the FGF signal pathway is crucial for cell proliferation activity in epithelial cells and is involved in the organization of the rugae, the thickened lines on the secondary palate.

Previous studies have reported the expression pattern of FGF ligands and their receptors during embryonic development. There are multiple FGF receptor isoforms owing to RNA splicing, and these isoforms have tissue-specific expression patterns. For instance, Fgfr2 IIIb is expressed in the epithelium and Fgfr2 IIIc is expressed in mesenchymal cells, although exceptions exist at different developmental stages (Kettunen et al., ’98; Rice et al., 2004). These observations suggest that Fgfr2b conventional knockout mice may be highly similar to epithelial tissue-specific conditional knockout mice. Surprisingly, K14-Cre;Fgfr2fl/fl and Fgfr2b-/- mice exhibit some phenotypes that differ as well as common defects in the eye and skin. First of all, the severity of the limb formation defect is significantly different. Fgfr2b-/- mice exhibited failure of limb initiation (De Moerlooze et al., 2000), whereas K14-Cre;Fgfr2fl/fl mice showed normal forelimb development and compromised hindlimb development. X-gal staining of K14-Cre;R26R mice demonstrates that Cre recombinase derived from the K14 promoter is active around E10.5 in the hindlimb but not in the forelimb, implying that epithelial cells in the forelimb escape the deletion of Fgfr2 at early developmental stages. This observation in K14-Cre;Fgfr2fl/fl mice is consistent with the previous finding that Fgfr2b plays a critical role in the apical endodermal ridge (AER) during limb formation (Revest et al., 2001).

Fgfr2b null mice exhibited tooth defects and cleft palate (De Moerlooze et al., 2000), but the mechanism of the tooth defect has remained unknown. In this study, we conclude that the tooth defect in the absence of Fgfr2 is likely the result of a cell proliferation defect in the dental epithelium. Although Fgf8 is not the ligand for Fgfr2 (Ornitz et al., ’96), Fgf8 expression is reduced in K14-Cre;Fgfr2fl/fl mice in the presumptive tooth region, implying that Fgf8 is a downstream target gene of FGF signaling through FGFR2. We have recently shown that the antagonistic interaction between BMP4 and Fgf8 and the setup of proximal vs. distal domains of the first branchial arch epithelium is dependent upon Smad4-mediated BMP signaling in the neural crest-derived mesenchyme (Ko et al., 2007). Interestingly, however, the induction of Fgf8 expression in the first branchial arch epithelium is not dependent on the neural crest cells. For example, Fgf8 expression is unaffected in Msx1 null or Msx1/Msx2 double null mice (Bei and Maas, ’98; Ishii et al., 2005). This study provides crucial evidence that FGF signaling through Fgfr2 is required for the induction of Fgf8 in the first branchial arch epithelium during craniofacial development. In spite of the reduction of Fgf8 expression, tooth development is normal until E12.5, implying there is likely compensation by other molecules. Pitx2 expression in K14-Cre;Fgfr2fl/fl mice is indistinguishable from control mice. Recently, it has been reported that Pitx2 cooperates with Lef-1 for activation of β-catenin during tooth initiation (Amen et al., 2007). These results suggest that members of the Wnt canonical pathway with Pitx2 might compensate for Fgf8 function during tooth initiation. A previous study of Fgfr2b null mice has reported that Fgf8 is not downstream of Fgfr2 IIIb isoform during limb development (Revest et al., 2001). Recently Lu and coworkers report that AER-specific knockout of all isoforms of Fgfr2 causes the reduction of Fgf8 during limb development, which is consistent with our observations (Lu et al., 2008). Furthermore, mice with epithelial-specific Fgfr2b gene deletion do not exhibit tooth retardation and cleft palate (Grose et al., 2007). Taken together, these observations imply that another isoform of Fgfr2 such as IIIc not only is involved in the induction of Fgf8 but also compensates for the function of Fgfr2 IIIb.

In regards to cleft palate, Fgf2b conventional knockout mice exhibit reduced cell proliferation defect in both epithelial and mesenchymal cells (Rice et al., 2004). Furthermore, the palatal shelves of Fgfr2b conventional knockout mice were shorter and did not show palatal projection lateral with the tongue. In contrast, we detect a cell proliferation defect only in the palatal epithelium in K14-Cre;Fgfr2fl/fl mice. Although the size of the palatal shelves in K14-Cre;Fgfr2fl/fl mice are smaller than control mice, palatal projection lateral to the tongue does occur. Taken together, these observations imply that there is an intrinsic requirement for FGF signaling in the epithelium and Fgfr2b expressed in the palatal mesenchyme is likely to be responsible for mediating palatal mesenchymal cell proliferation.

In summary, our epithelial-specific gene deletion of Fgfr2b shows that FGF signaling functions through FGFR2. Future exploration of the difference in the phenotypes of conventional and conditional knockout mice may provide insights into the reciprocal interactions of epithelial and mesenchymal cells during tooth and palate development.

ACKNOWLEDGMENTS

We thank Dr. Julie Mayo for critical reading of the manuscript, Drs. David Ornitz and Sarah Millar for Fgfr2 floxed and K14-Cre mice, respectively. This study was supported by grants from the National Institute of Dental and Craniofacial Research, NIH (DE012711 and DE014078) to Yang Chai.

Grant sponsors: The National Institute of Dental and Craniofacial Research; NIH; Grant numbers: DE012711; DE014078.

LITERATURE CITED

  1. Amen M, Liu X, Vadlamudi U, Elizondo G, Diamond E, Engelhardt JF, Amendt BA. PITX2 and beta-catenin interactions regulate Lef-1 isoform expression. Mol Cell Biol. 2007;27:7560–7573. doi: 10.1128/MCB.00315-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Andl T, Ahn K, Kairo A, Chu EY, Wine-Lee L, Reddy ST, Croft NJ, Cebra-Thomas JA, Metzger D, Chambon P, Lyons KM, Mishina Y, Seykora JT, Crenshaw EB, 3rd, Millar SE. Epithelial Bmpr1a regulates differentiation and proliferation in postnatal hair follicles and is essential for tooth development. Development. 2004;131:2257–2268. doi: 10.1242/dev.01125. [DOI] [PubMed] [Google Scholar]
  3. Avivi A, Yayon A, Givol D. A novel form of FGF receptor-3 using an alternative exon in the immunoglobulin domain III. FEBS Lett. 1993;330:249–252. doi: 10.1016/0014-5793(93)80882-u. [DOI] [PubMed] [Google Scholar]
  4. Beer HD, Vindevoghel L, Gait MJ, Revest JM, Duan DR, Mason I, Dickson C, Werner S. Fibroblast growth factor (FGF) receptor 1-IIIb is a naturally occurring functional receptor for FGFs that is preferentially expressed in the skin and the brain. J Biol Chem. 2000;275:16091–16097. doi: 10.1074/jbc.275.21.16091. [DOI] [PubMed] [Google Scholar]
  5. Bei M, Maas R. FGFs and BMP4 induce both Msx1-independent and Msx1-dependent signaling pathways in early tooth development. Development. 1998;125:4325–4333. doi: 10.1242/dev.125.21.4325. [DOI] [PubMed] [Google Scholar]
  6. Chai Y, Maxson RE., Jr Recent advances in craniofacial morphogenesis. Dev Dyn. 2006;235:2353–2375. doi: 10.1002/dvdy.20833. [DOI] [PubMed] [Google Scholar]
  7. Chai Y, Jiang X, Ito Y, Bringas P, Jr, Han J, Rowitch DH, Soriano P, McMahon AP, Sucov HM. Fate of the mammalian cranial neural crest during tooth and mandibular morphogenesis. Development. 2000;127:1671–1679. doi: 10.1242/dev.127.8.1671. [DOI] [PubMed] [Google Scholar]
  8. Chellaiah AT, McEwen DG, Werner S, Xu J, Ornitz DM. Fibroblast growth factor receptor (FGFR) 3. Alternative splicing in immunoglobulin-like domain III creates a receptor highly specific for acidic FGF/FGF-1. J Biol Chem. 1994;269:11620–11627. [PubMed] [Google Scholar]
  9. Cui XM, Shiomi N, Chen J, Saito T, Yamamoto T, Ito Y, Bringas P, Chai Y, Shuler CF. Overexpression of Smad2 in Tgf-beta3-null mutant mice rescues cleft palate. Dev Biol. 2005;278:193–202. doi: 10.1016/j.ydbio.2004.10.023. [DOI] [PubMed] [Google Scholar]
  10. De Moerlooze L, Spencer-Dene B, Revest J, Hajihosseini M, Rosewell I, Dickson C. An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in Mesenchymal–epithelial signalling during mouse organo-genesis. Development. 2000;127:483–492. doi: 10.1242/dev.127.3.483. [DOI] [PubMed] [Google Scholar]
  11. Grose R, Fantl V, Werner S, Chioni AM, Jarosz M, Rudling R, Cross B, Hart IR, Dickson C. The role of fibroblast growth factor receptor 2b in skin homeostasis and cancer development. EMBO J. 2007;26:1268–1278. doi: 10.1038/sj.emboj.7601583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Harada H, Toyono T, Toyoshima K, Yamasaki M, Itoh N, Kato S, Sekine K, Ohuchi H. FGF10 maintains stem cell compartment in developing mouse incisors. Development. 2002;129:1533–1541. doi: 10.1242/dev.129.6.1533. [DOI] [PubMed] [Google Scholar]
  13. Hogan BL. Morphogenesis. Cell. 1999;96:225–233. doi: 10.1016/s0092-8674(00)80562-0. [DOI] [PubMed] [Google Scholar]
  14. Hui CC, Joyner AL. A mouse model of greig cephalopolysyndactyly syndrome: the extra-toesJ mutation contains an intragenic deletion of the Gli3 gene. Nat Genet. 1993;3:241–246. doi: 10.1038/ng0393-241. [DOI] [PubMed] [Google Scholar]
  15. Indra AK, Li M, Brocard J, Warot X, Bornert JM, Gerard C, Messaddeq N, Chambon P, Metzger D. Targeted somatic mutagenesis in mouse epidermis. Horm Res. 2000;54:296–300. doi: 10.1159/000053275. [DOI] [PubMed] [Google Scholar]
  16. Ishii M, Han J, Yen HY, Sucov HM, Chai Y, Maxson RE., Jr Combined deficiencies of Msx1 and Msx2 cause impaired patterning and survival of the cranial neural crest. Development. 2005;132:4937–4950. doi: 10.1242/dev.02072. [DOI] [PubMed] [Google Scholar]
  17. Itoh N, Ornitz DM. Evolution of the Fgf and Fgfr gene families. Trends Genet. 2004;20:563–569. doi: 10.1016/j.tig.2004.08.007. [DOI] [PubMed] [Google Scholar]
  18. Kettunen P, Karavanova I, Thesleff I. Responsiveness of developing dental tissues to fibroblast growth factors: expression of splicing alternatives of FGFR1, -2, -3, and of FGFR4; and stimulation of cell proliferation by FGF-2, -4, -8, and -9. Dev Genet. 1998;22:374–385. doi: 10.1002/(SICI)1520-6408(1998)22:4<374::AID-DVG7>3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
  19. Ko SO, Chung IH, Xu X, Oka S, Zhao H, Cho ES, Deng C, Chai Y. Smad4 is required to regulate the fate of cranial neural crest cells. Dev Biol. 2007;312:435–447. doi: 10.1016/j.ydbio.2007.09.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Lu P, Yu Y, Perdue Y, Werb Z. The apical ectodermal ridge is a timer for generating distal limb progenitors. Development. 2008;135:1395–1405. doi: 10.1242/dev.018945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. McLeod MJ. Differential staining of cartilage and bone in whole mouse fetuses by alcian blue and alizarin red S. Teratology. 1980;22:299–301. doi: 10.1002/tera.1420220306. [DOI] [PubMed] [Google Scholar]
  22. Miki T, Bottaro DP, Fleming TP, Smith CL, Burgess WH, Chan AM, Aaronson SA. Determination of ligand-binding specificity by alternative splicing: two distinct growth factor receptors encoded by a single gene. Proc Natl Acad Sci USA. 1992;89:246–250. doi: 10.1073/pnas.89.1.246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Naski MC, Ornitz DM. FGF signaling in skeletal development. Front Biosci. 1998;3:d781–d794. doi: 10.2741/a321. [DOI] [PubMed] [Google Scholar]
  24. Nie X, Luukko K, Kettunen P. FGF signalling in craniofacial development and developmental disorders. Oral Dis. 2006;12:102–111. doi: 10.1111/j.1601-0825.2005.01176.x. [DOI] [PubMed] [Google Scholar]
  25. Ohuchi H, Hori Y, Yamasaki M, Harada H, Sekine K, Kato S, Itoh N. FGF10 acts as a major ligand for FGF receptor 2 IIIb in mouse multi-organ development. Biochem Biophys Res Commun. 2000;277:643–649. doi: 10.1006/bbrc.2000.3721. [DOI] [PubMed] [Google Scholar]
  26. Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol. 2001;2 doi: 10.1186/gb-2001-2-3-reviews3005. REVIEWS3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Ornitz DM, Xu J, Colvin JS, McEwen DG, MacArthur CA, Coulier F, Gao G, Goldfarb M. Receptor specificity of the fibroblast growth factor family. J Biol Chem. 1996;271:15292–15297. doi: 10.1074/jbc.271.25.15292. [DOI] [PubMed] [Google Scholar]
  28. Revest JM, Spencer-Dene B, Kerr K, De Moerlooze L, Rosewell I, Dickson C. Fibroblast growth factor receptor 2-IIIb acts upstream of Shh and Fgf4 and is required for limb bud maintenance but not for the induction of Fgf8, Fgf10, Msx1, or Bmp4. Dev Biol. 2001;231:47–62. doi: 10.1006/dbio.2000.0144. [DOI] [PubMed] [Google Scholar]
  29. Rice R, Spencer-Dene B, Connor EC, Gritli-Linde A, McMahon AP, Dickson C, Thesleff I, Rice DP. Disruption of Fgf10/Fgfr2b-coordinated epithelial–mesenchymal interactions causes cleft palate. J Clin Invest. 2004;113:1692–1700. doi: 10.1172/JCI20384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Sasaki T, Ito Y, Bringas P, Jr, Chou S, Urata MM, Slavkin H, Chai Y. TGFbeta-mediated FGF signaling is crucial for regulating cranial neural crest cell proliferation during frontal bone development. Development. 2006;133:371–381. doi: 10.1242/dev.02200. [DOI] [PubMed] [Google Scholar]
  31. Sasaki Y, O'Kane S, Dixon J, Dixon MJ, Ferguson MW. Temporal and spatial expression of Pax9 and Sonic hedgehog during development of normal mouse palates and cleft palates in TGF-beta3 null embryos. Arch Oral Biol. 2007;52:260–267. doi: 10.1016/j.archoralbio.2006.09.012. [DOI] [PubMed] [Google Scholar]
  32. Scotet E, Houssaint E. The choice between alternative IIIb and IIIc exons of the FGFR-3 gene is not strictly tissue-specific. Biochim Biophys Acta. 1995;1264:238–242. doi: 10.1016/0167-4781(95)00156-b. [DOI] [PubMed] [Google Scholar]
  33. Sekine K, Ohuchi H, Fujiwara M, Yamasaki M, Yoshizawa T, Sato T, Yagishita N, Matsui D, Koga Y, Itoh N, Kato S. Fgf10 is essential for limb and lung formation. Nat Genet. 1999;21:138–141. doi: 10.1038/5096. [DOI] [PubMed] [Google Scholar]
  34. Soriano P. Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat Genet. 1999;21:70–71. doi: 10.1038/5007. [DOI] [PubMed] [Google Scholar]
  35. Thesleff I, Sharpe P. Signalling networks regulating dental development. Mech Dev. 1997;67:111–123. doi: 10.1016/s0925-4773(97)00115-9. [DOI] [PubMed] [Google Scholar]
  36. Wilkinson DG. In situ hybridaization: a practical approach. Oxford University Press; Oxford, UK: 1998. [Google Scholar]
  37. Xu X, Han J, Ito Y, Bringas P, Jr, Urata MM, Chai Y. Cell autonomous requirement for Tgfbr2 in the disappearance of medial edge epithelium during palatal fusion. Dev Biol. 2006;297:238–248. doi: 10.1016/j.ydbio.2006.05.014. [DOI] [PubMed] [Google Scholar]
  38. Xu X, Han J, Ito Y, Bringas P, Jr, Deng C, Chai Y. Ectodermal Smad4 and p38 MAPK are functionally redundant in mediating TGF-b/BMP signaling during tooth and palate development. Dev Cell. 2008;15:322–329. doi: 10.1016/j.devcel.2008.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Yu K, Xu J, Liu Z, Sosic D, Shao J, Olson EN, Towler DA, Ornitz DM. Conditional inactivation of FGF receptor 2 reveals an essential role for FGF signaling in the regulation of osteoblast function and bone growth. Development. 2003;130:3063–3074. doi: 10.1242/dev.00491. [DOI] [PubMed] [Google Scholar]
  40. Zhang Y, Wang S, Song Y, Han J, Chai Y, Chen Y. Timing of odontogenic neural crest cell migration and tooth-forming capability in mice. Dev Dyn. 2003;226:713–718. doi: 10.1002/dvdy.10274. [DOI] [PubMed] [Google Scholar]

RESOURCES