abnormalities in cartilage and bone development in the apert · bones are formed by one of two...

12
3537 Introduction Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The neurocranium is composed of membranous bones that are formed by direct ossification, and the sutures between them comprise apposing osteogenic fronts separated by mesenchymal cells (Opperman, 2000). This developmental process differs from endochondral bone formation at the base of the skull and long bones because the latter has an intermediate cartilage anlagen. In humans and mouse, these calvarial sutures appear at approximately 18 weeks or embryonic day 15 (E15) of gestation, respectively, and remain open at birth to allow continued brain growth. In mice, the two major midline sutures are the interfrontal (corresponding to metopic in humans) between the frontal bones and the sagittal between the parietal bones. Sutures transverse to the midline include the coronal sutures between the frontal and parietal bones (see Fig. S1 in the supplementary material). The midline interfrontal suture is neural crest derived, whereas, at the midline sagittal suture, there are neural crest cells present between the mesodermally derived parietal bones. The coronal suture lies at the interface of the neural crest- derived frontal bones and the mesoderm of the parietal bones (Jiang et al., 2002). At these sutures, a proportion of these cells are recruited to differentiate into osteoblasts and make new bone (Cohen and Kreiborg, 1993; Opperman, 2000). A cartilaginous layer underlies the lower part of the parietal bone and coronal sutures proximate to the endochondral bones at the base of the skull (Iseki et al., 1999). By contrast, there is no compelling evidence that cartilage is normally formed at the Apert syndrome is an autosomal dominant disorder characterized by malformations of the skull, limbs and viscera. Two-thirds of affected individuals have a S252W mutation in fibroblast growth factor receptor 2 (FGFR2). To study the pathogenesis of this condition, we generated a knock-in mouse model with this mutation. The Fgfr2 +/S252W mutant mice have abnormalities of the skeleton, as well as of other organs including the brain, thymus, lungs, heart and intestines. In the mutant neurocranium, we found a midline sutural defect and craniosynostosis with abnormal osteoblastic proliferation and differentiation. We noted ectopic cartilage at the midline sagittal suture, and cartilage abnormalities in the basicranium, nasal turbinates and trachea. In addition, from the mutant long bones, in vitro cell cultures grown in osteogenic medium revealed chondrocytes, which were absent in the controls. Our results suggest that altered cartilage and bone development play a significant role in the pathogenesis of the Apert syndrome phenotype. Key words: Apert syndrome, Fibroblast growth factor receptor 2, Mouse, Cartilage, Bone, Mesenchyme, Neural crest, Skull, Suture, Craniosynostosis Summary Abnormalities in cartilage and bone development in the Apert syndrome FGFR2 +/S252W mouse Yingli Wang 1,4, *, Ran Xiao 1,4, *, Fan Yang 5 , Baktiar O. Karim 2 , Anthony J. Iacovelli 1,4 , Juanliang Cai 1,4 , Charles P. Lerner 6 , Joan T. Richtsmeier 4,7 , Jen M. Leszl 7 , Cheryl A. Hill 7 , Kai Yu 8 , David M. Ornitz 8 , Jennifer Elisseeff 5 , David L. Huso 2 and Ethylin Wang Jabs 1,3,4,† 1 Institute of Genetic Medicine, Department of Pediatrics, The Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, USA 2 Department of Comparative Medicine, The Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, USA 3 Department of Medicine and Plastic Surgery, The Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, USA 4 Center for Craniofacial Development and Disorders, The Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, USA 5 Department of Biomedical Engineering, The Johns Hopkins University, 3400 N. Charles Street, Clark 106, Baltimore, MD 21218, USA 6 Departments of Cell Biology/Microinjection and Microchemistry, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA 7 Department of Anthropology, The Pennsylvania State University, 409 Carpenter Building, University Park, PA 16802, USA 8 Department of Molecular Biology and Pharmacology, Washington University Medical School, 660 S. Euclid Avenue, St Louis, MO 63110, USA *These authors contributed equally to this work Author for correspondence (e-mail: [email protected]) Accepted 13 May 2005 Development 132, 3537-3548 Published by The Company of Biologists 2005 doi:10.1242/dev.01914 Research article Development and disease Development

Upload: others

Post on 12-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3537

Introduction

Bones are formed by one of two types of ossification processes,intramembranous or endochondral bone formation. Theneurocranium is composed of membranous bones that areformed by direct ossification, and the sutures between themcomprise apposing osteogenic fronts separated bymesenchymal cells (Opperman, 2000). This developmentalprocess differs from endochondral bone formation at the baseof the skull and long bones because the latter has anintermediate cartilage anlagen. In humans and mouse, thesecalvarial sutures appear at approximately 18 weeks orembryonic day 15 (E15) of gestation, respectively, and remainopen at birth to allow continued brain growth. In mice, the twomajor midline sutures are the interfrontal (corresponding tometopic in humans) between the frontal bones and the sagittal

between the parietal bones. Sutures transverse to the midlineinclude the coronal sutures between the frontal and parietalbones (see Fig. S1 in the supplementary material).

The midline interfrontal suture is neural crest derived,whereas, at the midline sagittal suture, there are neural crestcells present between the mesodermally derived parietal bones.The coronal suture lies at the interface of the neural crest-derived frontal bones and the mesoderm of the parietal bones(Jiang et al., 2002). At these sutures, a proportion of these cellsare recruited to differentiate into osteoblasts and make newbone (Cohen and Kreiborg, 1993; Opperman, 2000). Acartilaginous layer underlies the lower part of the parietal boneand coronal sutures proximate to the endochondral bones at thebase of the skull (Iseki et al., 1999). By contrast, there is nocompelling evidence that cartilage is normally formed at the

Apert syndrome is an autosomal dominant disordercharacterized by malformations of the skull, limbs andviscera. Two-thirds of affected individuals have a S252Wmutation in fibroblast growth factor receptor 2 (FGFR2).To study the pathogenesis of this condition, we generated aknock-in mouse model with this mutation. The Fgfr2+/S252W

mutant mice have abnormalities of the skeleton, as well asof other organs including the brain, thymus, lungs, heartand intestines. In the mutant neurocranium, we found amidline sutural defect and craniosynostosis with abnormalosteoblastic proliferation and differentiation. We notedectopic cartilage at the midline sagittal suture, and

cartilage abnormalities in the basicranium, nasalturbinates and trachea. In addition, from the mutant longbones, in vitro cell cultures grown in osteogenic mediumrevealed chondrocytes, which were absent in the controls.Our results suggest that altered cartilage and bonedevelopment play a significant role in the pathogenesis ofthe Apert syndrome phenotype.

Key words: Apert syndrome, Fibroblast growth factor receptor 2,Mouse, Cartilage, Bone, Mesenchyme, Neural crest, Skull, Suture,Craniosynostosis

Summary

Abnormalities in cartilage and bone development in the Apert

syndrome FGFR2+/S252W mouse

Yingli Wang1,4,*, Ran Xiao1,4,*, Fan Yang5, Baktiar O. Karim2, Anthony J. Iacovelli1,4, Juanliang Cai1,4,Charles P. Lerner6, Joan T. Richtsmeier4,7, Jen M. Leszl7, Cheryl A. Hill7, Kai Yu8, David M. Ornitz8,Jennifer Elisseeff5, David L. Huso2 and Ethylin Wang Jabs1,3,4,†

1Institute of Genetic Medicine, Department of Pediatrics, The Johns Hopkins University School of Medicine, 733 North Broadway,Baltimore, MD 21205, USA2Department of Comparative Medicine, The Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore,MD 21205, USA3Department of Medicine and Plastic Surgery, The Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore,MD 21205, USA4Center for Craniofacial Development and Disorders, The Johns Hopkins University School of Medicine, 733 North Broadway,Baltimore, MD 21205, USA5Department of Biomedical Engineering, The Johns Hopkins University, 3400 N. Charles Street, Clark 106, Baltimore, MD 21218,USA6Departments of Cell Biology/Microinjection and Microchemistry, The Jackson Laboratory, 600 Main Street, Bar Harbor,ME 04609, USA7Department of Anthropology, The Pennsylvania State University, 409 Carpenter Building, University Park, PA 16802, USA8Department of Molecular Biology and Pharmacology, Washington University Medical School, 660 S. Euclid Avenue, St Louis,MO 63110, USA*These authors contributed equally to this work†Author for correspondence (e-mail: [email protected])

Accepted 13 May 2005

Development 132, 3537-3548

Published by The Company of Biologists 2005

doi:10.1242/dev.01914

Research article Development and disease

Dev

elop

men

t

Page 2: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3538

superior part of the parietal bone and midline sutures of themembranous neurocranium.

In humans one of the most severe conditions involvingabnormal sutural development and skull growth is Apertsyndrome (Cohen, 2000). In infancy, a patent midline defect ofthe metopic and sagittal sutures, and synostosis (prematurefusion) of the coronal suture, are present. During childhood,the sagittal and lambdoid sutures become synostosed (Kreiborgand Cohen, 1990). Other abnormalities of the skeleton includesevere syndactyly, and internal organs are also affected, but lessfrequently (Table 1).

Remarkably, Apert syndrome is caused by a limited numberof mutations. Ninety-nine percent of reported cases have oneof two missense mutations in adjacent amino acids, Ser252Trpand Pro253Arg, of fibroblast growth factor receptor 2 (FGFR2)(Park et al., 1995; Wilkie et al., 1995). The first mutation,S252W, is more common, occurring in 67% of patients, andhas been proposed to be associated with more severecraniofacial anomalies, whereas the second mutation, P253R,may be associated with more severe syndactyly (Slaney et al.,1996; von Gernet et al., 2000). Both mutations affect the highlyconserved linker region between the immunoglobulin-like IIand III domains and result in increased affinity and alteredspecificity of fibroblast growth factor (FGF) ligand binding(Ibrahimi et al., 2001; Yu et al., 2000).

Normally, the ligand-binding characteristics of FGFR2 varydepending on its isoform. Alternative splicing of exons IIIb andIIIc encoding the immunoglobulin-like III domain results inspliceforms that are expressed predominantly by epithelial ormesenchymal cells, respectively (Miki et al., 1992; Orr-Urtregeret al., 1993). Distinct ligands bind to the IIIb and IIIc isoforms.Ligands FGF7 and FGF10 activate FGFR2 IIIb, whereas FGFs2, 4, 6, 8 and 9 activate FGFR2 IIIc. An Alu insertion flankingexon IIIc in an Apert syndrome patient was found to affectsplicing and cause the ectopic expression of FGFR2 IIIb infibroblasts from the patient (Oldridge et al., 1999).

FGFR2, as well as the other three FGFR tyrosine kinasesand their 22 FGF ligands, is known to play a crucial role in thecontrol of cell migration, proliferation, differentiation andsurvival, by activating two primary pathways, the mitogen-activating protein kinase pathway (Kouhara et al., 1997; Ornitzand Itoh, 2001) and protein kinase C pathway (Debiais et al.,2001; Lemonnier et al., 2001). Signaling through FGFR2regulates stem cell proliferation, affecting different lineagessuch as osteoblasts and chondroblasts (De Moerlooze et al.,2000; Eswarakumar et al., 2002; Iseki et al., 1999; Ornitz andMarie, 2002).

Chen et al. created a transgenic mouse with an Fgfr2+/S250W

mutation (Chen et al., 2003). (Although the correct conservedserine at position 252 was targeted, it was incorrectlydesignated as residue 250 – Chu-Xia Deng, personalcommunication.) Their mice were found to have severalfeatures similar to Apert syndrome, including a small bodysize, brachycephaly, midface hypoplasia, short presphenoidbone, wide-spaced eyes, and malocculsion. The long bonesrevealed few abnormalities. The heights of the growth platescorrelated with the smaller size of the mutants, and the columnof proliferating chondrocytes was slightly short in older mice(postnatal day 10, P10). Most importantly, Chen et al.performed detailed studies of the transverse coronal sutureswhere they found premature closure, decreased boneformation, and increased apoptosis, but no obvious change incell proliferation and differentiation. Thus, they suggest thatdysregulated apoptosis plays an important role in thepathogenesis of Apert syndrome-related phenotypes.

In this study, we explored the pathogenesis of other Apertsyndrome malformations, including the midline suturalabnormalities and those of the internal organs, by introducingthe Fgfr2 S252W mutation into the mouse genome. Ourresulting heterozygotes have phenotypic features consistentwith the human Apert syndrome. This mutation altersproliferation and differentiation of osteoblasts at the midlinecalvarial sutures. It is also responsible for ectopic cartilageformation in the neurocranium and cartilaginous abnormalitiesin several organs including the long bones. These findingshighlight the delicate balance of migration, proliferation anddifferentiation that orchestrate individual suture and bonedevelopment (Marie et al., 2002).

Materials and methodsGeneration of targeting construct and mutant mice

We used a 6.28 kb genomic DNA fragment containing exons IIIa, IIIband IIIc (exons 7, 8 and 9) of the murine Fgfr2 gene cloned intopBluescript SK– (Stratagene) to make a construct (Fig. 1A). A755_756CA>GG substitution, resulting in a Ser252Trp mutation atthe residue homologous to human FGFR2 amino acid 252, wasintroduced into exon IIIa using site-directed mutagenesis. We inserteda thymidine kinase (TK) gene cassette at the 5′ XhoI site upstream ofthe targeting DNA fragment and a neo cassette with flanking loxPrecognition sites into intron IIIa, oriented in an oppositetranscriptional direction to the target Fgfr2. The final targeting vectorof 13.6 kb was confirmed by sequencing, linearized by NotI digestion,and introduced into R1 ES cells by electroporation (carried out by TheJackson Laboratory).

We identified two clones with the targeted mutant allele, andgermline chimeric mice were generated from each clone (Fig. 1B).

Development 132 (15) Research article

Table 1. Phenotypic comparison between Apert syndromepatients and mice

Affected organ Humans* (%) Mice (%)

Skull 100 (68/68) 100 (10/10)Craniosynostosis 100 (68/68) 67 (6/9)‡

Midline suture 100† (N/A) 100 (10/10)Palate 58 (39/67) 100 (8/8)Central nervous system 21 (10/48) 20 (2/10)§

Trachea Eight cases† 8 (1/13)§

Thymus No reported cases 50 (5/10)Lungs 1.5† (2/136) 100 (13/13)Cardiovascular system 16 (11/67) 64 (7/11)Gastrointestinal system 1.5† (2/136) 50 (5/10)Genitourinary system 8† (10/123) 0 (0/10)Syndactyly 100 (68/68) 0 (0/13)Long bones¶ 95† (36/68) 90 (9/10)

*Frequency in humans based on patients with a FGFR2+/S252W mutation(Cohen and Kreiborg, 1993; Park et al., 1995; Slaney et al., 1996).

†Based on patients with either a +/S252W or a +/P253R mutation (Cohenand MacClean 2002).

‡Craniosynostosis based on abnormal histology in mice.§Central nervous system and trachea not examined by serial sections in

mice.¶Long bones refer to short humeri in humans and to abnormal histology in

mice.N/A, not available.

Dev

elop

men

t

Page 3: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3539Apert syndrome mice with cartilage changesDevelopment and disease

Male chimeras were mated with C57BL/6J females to achievegermline transmission of the mutant allele. The presence of the neocassette interfered with mutant allele expression. Therefore, F1heterozygotes with neo (+/S252Wflox) were crossed with CMV orEIIA promoter Cre transgenic C57BL/6J mice (CMV-Cre, TransgenicCore Facility of the Johns Hopkins University School of Medicine;EIIA-Cre, The Jackson Laboratory) to remove the neo cassette. Careand use of mice for this study were in compliance with the relevantanimal welfare guidelines approved by the Johns Hopkins UniversityAnimal Care and Use Committee.

Mutant allele genotyping and RT-PCR expression analysis

Genotypes of tail DNA from the resulting progeny were determinedby PCR analysis using primers from within Fgfr2 intron 6 (forwardprimer F, 5′-CTCGGAGTGTAACGTGTTC-3′), intron IIIa (reverseprimer R, 5′-CAACAGGAAATCAAAGACC-3′) and the neo cassette(forward primer Fn, 5′-GATGTTTCGCTTGGTGGTC-3′) (Fig.1A,C), and their identity was confirmed by DNA sequencing. Weidentified Fgfr2 mutant allele expression in total RNA isolated fromvarious organs and tissues from Fgfr2+/S252W mice by using theRNAqueousTM-4PCR Kit (Ambion) and the Omniscript RT Kit(QIAGEN) (Fig. 1D). RT-PCR amplification was performed usingprimers corresponding to the Fgfr2-coding sequence of exons 5 and10 (forward primer, 5′-CAACACCGAGAAGATGGAG-3′ and reverseprimer, 5′-CCATGCAGGCGATTAAGAAG-3′). Products weredigested with SfiI to distinguish the mutant from the wild-type allele.

Image analyses of the skull from the Fgfr2+/S252Wflox/S252W

mouse

Adult mice were euthanized by halothane inhalation and X-rays wereperformed. After removing the skin, mice were decapitated andthe heads were fixed in 4% paraformaldehyde. Micro-computedtomography scans were acquired by the Center for QuantitativeImaging at the Pennsylvania State University. Using eTDIPS software(http://www.cc.nih.gov/cip/software/etdips/), 3D reconstructionswere made using the surface reconstruction module of eTDIPS, and24 biological landmarks were located twice on each skull andmandible. The means of the three-dimensional coordinates of theselandmarks from each mouse were used to analyze morphologicaldifferences between mice. A quantitative comparison of the lineardistances estimated among the 24 landmarks using EuclideanDistance Matrix Analysis, or EDMA (http://oshima.anthro.psu.edu),provided precise measures of localized differences between mutantand control littermates.

Bone and cartilage staining, and measurements ofskeleton

Skeletal staining with Alizarin Red S and Alcian Blue was performed(McLeod, 1980). Skull measurements, including skull and nose lengths,skull height and width, mandibular and maxillary lengths, and innercanthal distance (Richtsmeier et al., 2000), and limb measurements,including the length of the humerus, radius, ulna, femur, tibia and fibula,were taken with digital calipers (Fisher Scientific).

Histological analysis and special staining of the skeleton

Whole heads of embryos at E14.5 to P1 were dissected. Histologicalsections (5 μm) were prepared from selected tissues that had beenfixed in 4% paraformaldehyde and embedded in paraffin. Sectionswere stained with Hematoxylin and Eosin (HE) for histology. Alkalinephosphatase (ALP, specific for osteoblasts) and tartrate resistant acidphosphatase (TRAP, specific for osteoclasts) in skull sutures andgrowth plates were stained using the TRACP and ALP double-stainkit (TaKaRa Bio).

Immunohistochemical and TUNEL assays of skeleton

Skull sutures, sectioned as above, were deparaffinized and hydratedthrough a xylene and graded alcohol series. The immunohistochemical

assays for Ki67 (Novocastra) and collagen II Ab-2 (Lab Vision) wereperformed using the VECTOR M.O.M Immunodetection Kit (VectorLaboratories). To assess cell proliferation, the slides were incubatedwith anti-Ki67 antibody, and visualized with biotinylated anti-mouseIgG (Vector) followed by the peroxidase reaction (brown color forKi67-positive nuclei). Cell proliferation was analyzed by counting thenumber of Ki67-positive cells in more than 100 cells from a definedarea, including and between the osteogenic fronts in three serialsections from four mutant and four control littermates for eachdevelopmental stage. The counts for Ki67-positive cells in mutant andwild-type embryos were compared by the t-test. To assess apoptosis,the TUNEL assay was performed, using the In Situ Cell DeathDetection Kit, POD (Roche Applied Science) for detection of apoptoticcell death by light microscopy.

In situ hybridization

In situ hybridization was performed on sections as described byWilkinson (Wilkinson, 1992) with modifications. The mouseosteonectin (ON), osteopontin (OP) and Sox9 cDNA fragments wereeach cloned into the pCR®II-TOPO® Vector. The plasmids werelinearized, and antisense and sense single-stranded RNA probes weregenerated with the T7 and SP6 RNA polymerase.

In vitro culture

Cells were obtained from limbs of newborn Fgfr2+/S252W mutant andwild-type mice. The long bones were dissected and connective tissuewas removed. The bone samples from the middle third of the shaftwere washed and centrifuged with phosphate-buffered saline (PBS,GIBCO). The samples were then digested in 1 mg/ml sterilecollagenase D (Boehringer Mannheim) solution at 37°C for 2 hours,centrifuged and washed with PBS before resuspension in standardculture medium [Dulbecco’s modified Eagle’s medium (DMEM;GIBCO), 10% fetal bovine serum, 100 unit/ml penicillin and 100μg/ml streptomycin]. Cells were plated onto a flask and mediumchanges occurred first after 3 days to allow cell attachment and thenthree times a week until confluency. Expanded cells were then placedin 3D culture (Elisseeff et al., 2005) and incubated in standardosteogenic medium for 3 weeks to evaluate differentiation. Osteogenicmedium consisted of high-glucose DMEM, 100 nM dexamethasone(Sigma-Aldrich Co.), 50 µg/ml ascorbic acid 2-phosphate, 10 mM β-glycerophosphate, 10% fetal bovine serum, 100 unit/ml penicillin and100 μg/ml streptomycin. Medium was changed every 2-3 days. At theend of 3 weeks in culture, wet and dry weights from all constructs fornormalization of extracellular matrix content were obtained after 48hours of lyophilization. ALP and calcium quantification, Von Kossastaining for mineralization (Pittenger et al., 1999), andimmunohistochemical analyses for collagen type I and II (ResearchDiagnostics, rabbit polyclonal antibodies), were performed. Forquantitative ALP assay, tissue cultures or constructs werehomogenized in 0.75 M 2-amino-2-methyl propanol (Sigma; pH 10.3)solution and the supernatants were collected for ALP assay usingSigma ALP Determination Kits (Sigma Diagnostics 245), followingthe manufacturer’s protocol. For quantitative calcium measurement,lyophilized tissue constructs were homogenized in 0.5 M HCl andvigorously vortexed for 16 hours at 4°C. The supernatant wascollected for calcium assay by following the manufacturer’s protocol(Sigma Diagnostics 587). Three samples from each group wereharvested for each assay.

Results

Generation of Fgfr2+/S252W mutant mice

We designed a gene-targeting construct to knock-in the humanApert FGFR2 point mutation, site specifically, generating thehomologous S252W amino acid change in the mouse (Fig. 1).

Dev

elop

men

t

Page 4: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3540

With the neo cassette, the mutant allele (S252Wflox) washypomorphic because it was either not expressed or itsexpression was reduced as determined by RT-PCR (see TableS1 in the supplementary material). Upon mating these micewith Cre transgenic mice, the neo gene was removed andresulting offspring expressed the mutant S252W allele inmultiple organs and tissues, including the skull, sagittal andcoronal sutures, palate, mandible, limbs, brain, thymus, heart,lung, liver, kidney, stomach and intestine (Fig. 1D, expressionin skull and lung is shown). Heterozygous Fgfr2+/S252W mutantmice originating from two independently derived ES cellclones were born at the expected frequency of 25% and hadthe same phenotypic features. At birth they were smaller, andtheir weight was 83% that of their wild-type littermates[+/S252W (n=20) versus +/+ (n=112): 1.225±0.091 g versus1.485±0.180 g, P<0.001]. Neonatal lethality occurreduniformly within 24 to 36 hours after birth due to apparentrespiratory failure with palatal abnormalities, lung atelectasisand evidence of aerophagia. Although most humans with Apertsyndrome survive the early postnatal period, a subset has beendescribed with early postnatal death, thickened nasal cartilage,palate shape abnormalities, complete tracheal sleeve, and lungabnormalities (Cohen, 2000).

Skeletal and other organ abnormalities inFgfr2+/S252W mutant miceAutopsy with skeletal preparations of mutant mice revealedmultiple malformations that resembled those of human Apertsyndrome (Fig. 2). A total of 33 Fgfr2+/S252W mutant mice wereanalyzed at P1 (Table 1). The skull length, skull height andupper jaw length were significantly reduced when comparedwith control littermates [+/S252W (n=4) versus +/+ (n=4):average skull length 9.415 mm versus 10.450 mm, P=0.006;skull height 6.135 mm versus 6.570 mm, P=0.028; upper jawlength 5.399 mm versus 6.200 mm, P=0.018]. Their skullwidth, nose length, inner canthal distance, and lower jawmeasurements did not differ from those of controls (P=0.052to 0.842).

Other skeletal system abnormalities included increasedcartilage of the basicranium, malformation of the palate,thickened nasal cartilage, fusion of joints separating thezygomatic arch bones, fusion of bones of the sternum, andcomplete cartilage sleeve of the trachea (Fig. 2). Although themutant mice were smaller, the upper and lower limb lengthswere proportional between mutant and control littermates(ratio of humerus/radius in +/S252W versus +/+: 0.836 versus0.838, P=0.975; ratio of femur/tibia in +/S252W versus +/+:

Development 132 (15) Research article

Fig. 1. Generation of the targeting constructand Fgfr2+/S252W mice. (A) Structure of aportion of the wild-type Fgfr2 gene with exonsIIIa, IIIb, IIIc and 10; the targeting constructwith the TK cassette and neo cassette withflanking loxP sequences; and the mutant alleleproduced by homologous recombination andwith neo deletion mediated by Cre. The755_756CA>GG, S252W mutation (*) wasintroduced into exon IIIa. Probes (–) andrestriction enzymes (St, StyI; H, HindIII; S,SacI) used for Southern blot analysis, and PCRprimers (F, Fn, R; arrowheads) used forgenotyping are shown. (B) Identification ofmutant and wild-type alleles in twoindependent ES cell clones using Southern blotanalysis with 5′ and 3′ probes. (C) Genotype ofa litter from +/S252Wflox�+/Cre by PCR of tailDNA. Lanes 1, 3 and 5: heterozygote with+/S252Wflox, showing wild-type allele (521 bp)and hypomorphic allele with neo (1.1 kb).Lanes 2 and 4: heterozygote mutant +/S252W,showing mutant allele after neo deletion (581bp). Lane 7: hypomorphic mutant+/S252Wflox/S252W. Lanes 6 and 8: wild type(+/+). (D) RT-PCR detection of mutanttranscript in lung and skull tissues. Othertissues tested expressed the mutant allele (datanot shown). The mutation introduces anadditional SfiI restriction site. (E) Grossappearance of +/S252W and wild-typepostnatal day (P)1 mice. Note the small bodysize of the mutant mouse. The difference ofbody weight between the mutant and controlmice was significant.

Dev

elop

men

t

Page 5: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3541Apert syndrome mice with cartilage changesDevelopment and disease

0.616 versus 0.595, P=0.790). None of the mutants exhibitedsyndactyly.

Histopathological analysis of the brain revealed mildhydrocephalus with enlarged ventricles (Fig. 2). In the lungs,moderate and diffuse atelectasis with lack of alveolarexpansion and the presence of proteinaceous fluid in the alveoliwere observed. Multifocal and acute bronchiolectasis withmild dilation of the bronchioles were found. Within the thymusthere was diffuse lymphoid necrosis and apoptosis of varyingseverity (data not shown). Cardiovascular abnormalitiesincluded dilation of the atria and great vessels at the base ofthe heart, and mild luminal ectasia was found in the stomachand the intestine, consistent with aerophagia (data not shown).

Skull abnormalities in the Fgfr2+/S252Wflox/S252W

mutant mice

We analyzed the only two mutant mice with the hypomorphicS252Wflox allele that survived to adulthood. In these mice, theneo cassette was not completely removed in all tissues (Fig.1C, lane 7). The expression levels of both mutant Fgfr2 IIIb

and IIIc alternative transcripts in hypomorphic mutant micewere generally reduced by 16% to 41%, when compared withthose of the normal transcripts in multiple tissues examined,including lung, liver, kidney and stomach (see Table S1 in thesupplementary material). At 10 months, these mutants had anabnormal head shape, with a body weight that was 29%(mutant 10.63 g versus +/+ 36.35 g) and a length that was 68%(nasal tip to base of tail, mutant 62.69 mm versus +/+ 92.27mm) of those of their littermate controls (Fig. 3). Micro-CT ofthe adult skull revealed multiple abnormalities with aninterfrontal sutural defect (Fig. 3C, arrow), dysmorphology ofthe nasal bones, and obliteration of the fronto-premaxillary,premaxillary-maxillary, and nasal-frontal sutures. Thestructures of the face and palate were 40-50% smaller in themutant mouse than in the control mouse, while structures ofthe neurocranium and basicranium were reduced by 30-40%.Linear distances measuring the width of the skull were affectedless than the length was, resulting in a very brachycephalicskull shape. The frontal process of the maxilla showed anabnormally large sinus and bowing of the medial wall and the

Fig. 2. Gross bony structuresand histology of P1Fgfr2+/S252W mice.(A,C,E,G,I,K,M,O,Q,S) Controllittermates;(B,D,F,H,J,L,N,P,R,T)corresponding regions in mutantmice. (A-H) Alizarin Red S andAlcian Blue staining of (A-F)the skull and (G,H) the chest.(I-R) HE staining of the (I,J)brain, (K,L) palate, (M,N)trachea, (O,P) lung, and (Q,R)growth plates of long bone.(S,T) TRAP staining for growthplates. The mutant mice have: awide interfrontal suture (B,arrows define the width of thesuture); a shortened nasal snout(D, arrow); fusion of the jointsbetween the zygomatic archbones (F, arrows); a sternumwith abnormal fusions and bifidxiphoid process (H, arrow);hydrocephalus with enlargedventricles (J, arrows); a palatedefect (L, arrow); a completecartilage sleeve of the tracheawith abnormal cartilagethickening (N); atelectasis ofthe lungs with alveolarproteinaceous fluid (P);chondrocyte hypertrophy anddisorganization with irregularand thickened ossifyingcartilaginous spicules in thegrowth plates of long bone (R);and decreased osteoclasts at thechondro-osseous junction (T; S,arrow shows normal osteoclastsstained red). LV, lateral ventricle; 3V, third ventricle; TC, tracheal cartilage; P, zone of chondrocyte proliferation; H, zone of hypertrophy;M, zone of mineralization; O, zone of ossification at the chondro-osseous junction. Scale bars: A-H, 1 mm; I-T, 50 μm.

Dev

elop

men

t

Page 6: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3542

zygomatic process of the maxilla was more bowed laterally.The interparietal bone was compressed superiorly at thelambdoid suture whereas the parietal bones showed an obvioussuperior bulge. The inner surface of the calvarium showed‘thumb printing’ or fine indentations presumed to be secondaryto increased intracranial pressure. The mandible was very smallwith a dysmorphic angular process.

Sutural dysmorphology in Fgfr2+/S252W mutant mice

Sutures were examined microscopically to further characterizethe abnormalities observed in the mutant skulls during earlymouse development. Individual sutures exhibited differenthistological findings. The midline sagittal suture was similar inboth mutant and control littermates at E16.5. In E18.5 mutants,the space between the two osteogenic fronts of the sagittalsuture was reduced with increased cellularity. In P1 mutants,the osteogenic fronts were very proximate, and osteoid hadbegun to be deposited between them prior to synostosis of theparietal bones; by contrast, in the control littermates, theosteogenic fronts were widely separated (Fig. 4A-F).

The midline interfrontal suture showed an obvious gapbetween the osteogenic fronts of the frontal bones in the mutantmice when compared with control littermates (Fig. 4G,H). Thewidely separated osteogenic fronts correlate with the patentmidline defect observed macroscopically in skeletalpreparations (Fig. 2B) and CT scans of mutant mice (Fig. 3C,see arrow). These results in mice are consistent with themidline sutural defect found in all Apert syndrome infants(Kreiborg and Cohen, 1990).

The transverse coronal (Fig. 4I,J) and lambdoid sutures(Fig. 4K,L) in P1 mutants showed synostosis with osteoiddeposition. Our results are consistent with the coronalsynostosis found in P1 to P18 Apert syndrome mutant micereported by Chen et al. (Chen et al., 2003) and on CT scans ofApert syndrome patients (Kreiborg and Cohen, 1990).

Abnormal osteoblastic proliferation anddifferentiation at the midline sutures in mutant mice

The effects of the Fgfr2 +/S252W mutation on proliferation,differentiation and apoptosis were investigated at the sagittalsuture from E16.5 to P1. Cell proliferation was investigated bycounting Ki67-positive cells from a designated area in thesagittal sutures using serial sections. At E16.5, no differencein the number or distribution of Ki67-positive cells wasapparent between mutants and controls (data not shown). AtE18.5 and P1, there were twice as many proliferating Ki67cells at the sagittal suture in mutants (E18.5 +/S252W versus+/+: 98.0±5.0 positive cells versus 45.5±10.6 positive cells,P<0.023; P1 94.5±10.6 versus 26.5±0.7, P<0.018; Fig. 5A,B).These mutant Ki67-positive cells were abnormally distributedin the sutural space between the osteogenic fronts, as well asbeing localized at the fronts where they are present in controls.

Osteoblast differentiation was assessed using osteonectin(ON), osteopontin (OP) and alkaline phosphatase (ALP)markers. At E16.5, no differences were apparent in theexpression of these markers between mutants and controls(data not shown). At E18.5, ON, OP and ALP were expressedin the developing bone and in cells extending from the margin

Development 132 (15) Research article

Fig. 3. Skeletal features ofhypomorphic Fgfr2+/S252Wflox/S252W

mice. (A) The gross appearanceshows the smaller body size of themutant mouse at 8 weeks of age.(B) X-ray of the whole body at 8weeks of age shows short nasalbones and maxillary bones withoverriding lower incisors.(C) Micro-CT of the skull of a 10-month-old Fgfr2+/S252Wflox/S252W

mouse (right) shows extremebrachycephaly, diminutive andcurved nasal bones, extremereduction anteroposteriorly of thefrontal bones, obliterated fronto-nasal, fronto-premaxillary andmaxillary-premaxillary sutures,and a defect of the interfrontalsuture in the skull (arrow).

Dev

elop

men

t

Page 7: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3543Apert syndrome mice with cartilage changesDevelopment and disease

of the bone plates into the mid-sutural area in mutants (Fig.5C-H). These results suggest abnormal osteoblasticdifferentiation, because this latter area normally containsmesenchymal or osteoprogenitor cells, but no preosteoblasts.Apoptosis at the sagittal suture was studied by TUNELstaining. There was no apparent difference in apoptosis

between mutants and controls at E16.5, E18.5 and P1 (Fig. 5I,J;E18.5 is shown).

We also examined proliferation, differentiation andapoptosis at the midline interfrontal suture. In mutants, therewas a significant increase in the number of Ki67-positive cellsbetween the osteogenic fronts (E18.5 +/S252W versus +/+:181.0±4.2 positive cells versus 111.0±8.5 positive cells,P<0.01). The mutant Ki67 cells had a pronounced alteration intheir distribution. Rather than being enriched at the osteogenicfronts, Ki67-positive cells in the mutant extended from thewidely separated margins of the osteoid plates almost into themid-sutural space (Fig. 6C,D). In situ hybridization with ONand OP revealed an obvious lack of differentiating osteoblastsbetween the osteogenic fronts in mutants (Fig. 6E-H). Thisdifference could be detected at E16.5 and became moreobvious at E18.5. Similar to our results at the sagittal suture,there was no obvious difference in apoptosis at the interfrontalsuture of mutants and controls from E16.5 to E18.5 (data notshown).

Abnormal cartilage at the midline sagittal suture inmutant mice

In Fgfr2+/S252W mice, there was ectopic cartilage at the sagittalsuture (n=20 mutants, n=26 controls; Fig. 7). Normally, withintramembranous bone formation at the sagittal suture, there isno intermediate cartilaginous stage. At E16.5, cartilage wasdetected consistently throughout most of the entire length ofthe sagittal suture of mutants. At E18.5, cartilage was presentalong more than half of the length of the sagittal suture(0.45±0.11 mm of 0.71± 0.05 mm, as determined by 5 μmserial sections), beginning at the junction between the parietaland interparietal bones (see Fig. S1 in the supplementarymaterial). At P1, the cartilage resembled that at E18.5 in thesagittal suture of mutants. Two chondrogenic markers, Sox9and collagen type II, were expressed in these cells (Fig. 7E-L).No chondrocytes or cartilage was detected histologically, andno Sox9 expression was present in mutant sagittal sutures atearlier stages of E14.5 or E15.5 (data not shown) or in controlsfrom E14.5 to E18.5. At the later P1 stage, cartilage wasdetected near the junction of the parietal and interparietal bonesat the sagittal suture in only one out of the 26 controls.

Abnormal cartilage in the limb of mutant mice

The long bones were analyzed by histopathology. There wasdisorganization of the growth plates with subtle irregularityof the hypertrophic zone and more prominent cartilage

Fig. 4. Histological analysis of the calvarial sutures in Fgfr2+/S252W

mice. (A,C,E,G,I,K) Control littermates; (B,D,F,H,J,L)corresponding regions in mutant mice. (A-F) HE staining shows thedevelopment of the sagittal suture from E16.5 to P1. (A,B) Noobvious difference of the sagittal sutures between the mutant andcontrol at E16.5. (C,D) More proximate osteogenic fronts are evidentat the mutant sagittal suture at E18.5. (E,F) Osteoid deposition isseen across the mutant sagittal suture at P1. (G-L) HE stainingshows: a wide gap between the osteogenic fronts at the mutantinterfrontal suture at P1 (G,H); presynostosis/synostosis with osteoiddeposition at the mutant coronal suture at P1 (I,J); andpresynostosis/synostosis at the mutant lambdoid suture at P1 (K,L).Arrows, osteogenic fronts; arrowheads, presynostosis/synostosis withosteoid formation; s, skin; p, parietal bone; b, brain; f, frontal bone;ip, interparietal bone. Scale bars: A-H,K,L, 50 μm; I,J, 25 μm.

Dev

elop

men

t

Page 8: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3544

mineralization at P1. Markedly decreased osteoclasts at thechondro-osseous junction were observed by TRAP staining(Fig. 2Q-T). Samples were isolated from the long bones ofmutant mice to obtain further evidence of abnormal cartilage

formation. Before culture, both mutant and wild-type cellsrepresented a heterogeneous population. They were positive forbone-related proteins (ALP, collagen type I) and cartilage-related protein collagen type II (see Fig. S2 in thesupplementary material). Then these cells were cultured inestablished osteogenic medium. After three weeks in culture,both mutant and wild-type cells still generated bone-relatedproteins ALP and Col I. Von Kossa staining showedmineralization in the pericellular regions, which was alsoconfirmed by quantitative calcium assays (+/S252W versus+/+: 4.25±0.38% versus 3.66±0.67% dry weight).Chondrocytes were detected by collagen type II staining ofmutant cells cultured in 3D osteogenic conditions, but wereabsent in control cells (Fig. 8E,F). The DNA content did notdiffer between mutant and wild-type cultures assayed at 1 dayand 21 days of growth, suggesting that increased proliferationmay not be the explanation for the presence of mutant

Development 132 (15) Research article

Fig. 5. Proliferation, differentiation and apoptosis at the midlinesagittal suture in Fgfr2+/S252W mice at E18.5.(A,B) Immunohistochemical staining of Ki67 shows increasednumbers and abnormal distribution of positive cells indicatingabnormal proliferation in mutants (arrows). (C-H) Expression ofosteogenic markers shows increased or abnormal differentiation inmutants. (C,D) Osteonectin in situ hybridization (arrows). (E,F) ALPstaining (arrows). (G,H) Osteopontin in situ hybridization (arrows).(I,J) TUNEL staining does not show an obvious difference inapoptosis between mutants and controls. (A,C,E,G,I) Controllittermates; (B,D,F,H,J) corresponding regions in mutant mice. s,skin; p, parietal bone; b, brain. Scale bars: A-J, 50 μm.

Fig. 6. Proliferation and differentiation at the midline interfrontalsuture in Fgfr2+/S252W mice at E18.5. (A,B) HE staining shows a gapin mutants. (C,D) Ki67 staining shows abnormal distribution ofpositive cells in the mid-sutural mesenchyme in mutants.(E-H) Expression of osteogenic markers shows displaced, decreasedor delayed differentiation in mutants. (E,F) Osteonectin in situhybridization; (G,H) osteopontin in situ hybridization.(A,C,E,G) Control littermates; (B,D,F,H) corresponding regions inmutant mice. Arrows, osteogenic fronts; s, skin; f, frontal bone; b,brain. Scale bars: A-H, 50 μm.

Dev

elop

men

t

Page 9: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3545Apert syndrome mice with cartilage changesDevelopment and disease

chondroctyes in culture (see Fig. S3 in the supplementarymaterial). Perhaps, the osteogenic medium does not support thegrowth of normal chondrocytes and does not inhibit that ofmutant cells.

Discussion

Apert syndrome mouse model

Using a Cre-mediated knock-in method, we have generatedmice carrying the S252W mutation, which is the most commonFGFR2 mutation associated with Apert syndrome in humans.The mutant mice serve as a model for Apert syndrome, as theirphenotype is consistent with the human condition (Table 1).Fgfr2+/S252W mice are neonatal lethal, small in size, anddemonstrate significant anomalies, including craniosynostosisand multiple visceral anomalies, as were evident by ourhistopathological studies. A high penetrance of affected organsin this mouse model indicates that signaling through Fgfr2 iscrucial to the normal development of the skeleton, as well asaffecting the brain, lungs, thymus, and other visceral organs.Survival of the hypomorphic, Fgfr2+/S252Wflox/S252W mutantmice makes them useful in studying the postnatal skullabnormalities related to this condition.

We confirmed the results of Chen et al. (Chen et al., 2003),including the presence of coronal synostosis in mutant mice.In addition, our analysis focused on the midline sagittal andinterfrontal sutures, where a skull defect is present in all humanApert syndrome newborns. Although, severe syndactyly is a

key feature of Apert syndrome in humans, none of the mutantmice from either laboratory exhibited syndactyly by grossinspection and histopathology. The absence of this feature isconsistent with the possibility that this mutation causes lesssevere digital abnormalities in humans when compared withthe Apert FGFR2 P253R mutation (Slaney et al., 1996; vonGernet et al., 2000). Others speculate that there is no humanphenotypic difference for these two mutations (Park et al.,1995). Thus, the absence of syndactyly in our mice may be aconsequence of species-specific modifications or of alternativesplicing affecting levels of expression of receptors (Oldridgeet al., 1999).

Abnormal bone development at the midline calvarialsutures in Apert syndrome

Previously in the literature there has been few, if any,histopathological studies of the midline sutures, especially theinterfrontal suture, in craniosynostosis mouse models. For theFgfr2+/S252W mutant, the interfrontal suture had increasedproliferation, but abnormally localized, decreased or delayeddifferentiation of osteoprogenitor cells to preosteoblasts.Therefore, abnormal proliferation and differentiation ofosteoblasts, not increased apoptosis, may underlie the metopicsutural defect in Apert syndrome.

At the mutant sagittal suture, the number of proliferating cellsand the expression of osteogenic markers were mislocalized andincreased, suggesting that the observed synostosis results fromincreased proliferation and abnormal differentiation. In mutant

Fig. 7. Abnormal cartilage at the midline sagittal suture in Fgfr2+/S252W mice from E16.5 to E18.5. (A-D) HE staining, (E-H) Sox9 in situhybridization, and (I-L) collagen type II immunohistochemical staining, showing the abnormal cartilage in mutants from E16.5 (left panels) toE18.5 (right panels). (A,E,I,C,G,K) Control littermates; (B,F,J,D,H,L) corresponding regions in mutant mice. Arrows, abnormal cartilage; s,skin; p, parietal bone; b, brain. Scale bars: A-L, 50 μm.

Dev

elop

men

t

Page 10: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3546

mice, the osteogenic markers are expressed abnormally in cellsextending from the margin of bone plates to the mid-suturalarea. It is possible that the Fgfr2 +/S252W mutation may causeabnormal differentiation at the developing sagittal sutures byaltering the fate of mesenchymal cells.

Although there is increased and abnormal proliferation atboth mutant midline sutures, we observed significantdifferences in their development. The abnormal differentiationat the sagittal suture leads to synostosis between the parietalbones, while at the interfrontal suture it results in the gapbetween the frontal bones. By contrast, in the mutant coronalsuture, there is no increased proliferation or differentiation, butincreased apoptosis that leads to synostosis (Chen et al., 2003).Our findings demonstrate that bone development in Apertsyndrome can vary among specific sutures and even suturalsynostoses can be due to either increased growth byproliferation and differentiation, or decreased growth withapoptosis across a suture.

Abnormal cartilage development in Apert syndrome

We provide strong evidence that abnormal cartilage formationis involved in the pathogenesis of Apert syndrome. In humans,

although cartilage abnormalities have been reported in thiscondition, as well as in other craniosynostosis syndromes suchas Crouzon and Pfeiffer (Cohen and Kreiborg, 1992; Cohenand Kreiborg, 1993; Kreiborg et al., 1993; Kreiborg et al.,1999), abnormal cartilage formation has not previously beenrecognized as being significant. In our mouse model, the Fgfr2+/S252W mutation has a generalizable effect on thedevelopment of cartilage. Chondrogenic markers and increasedcartilage were found in the mutant sagittal suture at embryonicstages when normally they are not present. These findingssuggest that either there was a significant increase in cartilageproliferation at the junction of the parietal and interparietalbones, where it normally may be present, or that there isectopic cartilage formation from progenitor cells. We alsoobserved increased cartilage at the basicranium, nasalturbinates, trachea and long bones of our mutant mice. At thelong bone growth plates, abnormal cartilage formation wasdetected histologically in vivo, and this was further supportedby our in vitro cultures. These results indicate that the Fgfr2+/S252W mutation significantly enhances the formation ofcartilage in some organs, and we speculate that abnormalchondrogenesis may occur.

Interestingly, our results differ for the two midline sutures.At the sagittal suture, there is ectopic cartilage and synostosis,and at the interfrontal suture there is a defect where no cartilagewas found. The distinct tissue origins of the sagittal andinterfrontal sutures may reflect their mutant osteogenicpotentials. The sagittal suture is formed from neural crest cellsand mesoderm, whereas the interfrontal suture is neural crestderived (Jiang et al., 2002). The cranial neural crest cells cangive rise to either cartilage or bone (Le Douarin et al., 1993;Noden, 1983; Selleck et al., 1993). In vitro studies have shownthat mutant FGFRs can induce ectopic cartilage frompremigratory neural crest cells early in development. Petiot etal. demonstrated that transfection of activating FGFR1 K656Eor FGFR2 C278F mutations can induce cartilagedifferentiation when electroporated into quail premigratoryneural crest cells (Petiot et al., 2002), but this effect isdrastically reduced if transfection is carried out after the onsetof neural crest migration. Therefore, the observed abnormaldevelopment of cartilage in Apert syndrome may be due to themis-migration or mis-positioning of neural crest cells at anearly embryonic stage.

Of note, there have been previous reports of abnormalcartilage formation in the sagittal suture in pathologic states.In mutant mice overexpressing Msx2, cartilage has beendemonstrated to underlie the midline sagittal suture (Liu et al.,1999). Mice, exposed to retinoic acid at E10, develop cartilagein the parietal region concomitant with the decrease in theintramembranous ossification (Jiang et al., 2002), suggestingthat neural crest or mesenchymal cell fate may be sensitive toMsx2 dosage. Rice et al. reported that cartilaginous rods areoccasionally and transiently seen in the sutural mesenchymearound the time of birth (Rice et al., 2000), and this may be atissue reaction to mechanical irritation.

The complexity in the precise temporospatial definition ofeach developing suture in Apert syndrome is underscored bythe variable effects of the Fgfr2 +/S252W mutation, differencesin tissue origins, and potential genetic and environmentalmodifiers. These distinctions cannot be easily studied in humansurgical or cultured cells because of the lack of specimens at

Development 132 (15) Research article

Fig. 8. Chondrocytes in cultured cells from the limb of P1Fgfr2+/S252W mice. (A,B) Von Kossa staining at the pericellularregions and (C,D) collagen type I immunohistochemical staining aresimilar for both mutants and controls. (E,F) Collagen type II stainingis positive for the mutants, but is absent in controls. (A,C,E) Controllittermates; (B,D,F) mutants. Scale bars: A-F, 100 μm.

Dev

elop

men

t

Page 11: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3547Apert syndrome mice with cartilage changesDevelopment and disease

different developmental stages, sites, and controls. In fact,previous studies on human samples have differing results.Lomri et al. (Lomri et al., 1998) and Lemonnier et al.(Lemonnier et al., 2000) concluded that there was increasedosteoblast differentiation of calvarial cells without making adistinction between sagittal, metopic and coronal synostosedsutures in Apert syndrome fetuses and infants. Mansukhani etal. (Mansukhani et al., 2000) noted an opposite effect ofinhibition of differentiation, and increased apoptosis inimmortalized human osteoblasts, not specifically derived fromsutures, introduced with the FGFR2 S252W mutation.

Insights into the mechanism of the Fgfr2 +/S252Wmutation

Mutant mice with loss of function or dominant-negativemutations in Fgfr2 showed abnormalities in some of the sameorgans that are affected in our Apert syndrome mouse model.Mice homozygous for null alleles of Fgfr2 die at E10.5 withmultiple defects in organogenesis, and chimeras between thismutant and wild-type had abnormal epithelial-mesenchymalinteractions, respiratory failure and intramembranous andendochondral bone formation (Arman et al., 1998; Arman etal., 1999; Xu et al., 1998). A mouse with a specific deletionand a dominant-negative loss of Fgfr2 IIIb isoform functionresulted in craniofacial (wide cleft palate, reduced maxillarybone, absent otic capsule, rudimentary inner ear structures) andtruncating limb anomalies, dysgenesis of several visceralorgans (thymus, glandular stomach, pancreas, kidney), andagenesis of the tooth bud, salivary gland, thyroid, pituitary andlung (Celli et al., 1998; De Moerlooze et al., 2000).

Mice created by a specific deletion of Fgfr2 IIIc functionassociated with exon switching more closely resemble ourApert syndrome mice. Hemizygotes were affected withneonatal growth retardation and death, coronal synostosis,ocular proptosis with fused joints of the zygomatic arch bones,precocious sternal fusion, and abnormalities in secondarybranching in several organs, such as the lungs, kidneys andlacrimal glands (Hajihosseini et al., 2001). With a splice-switchmechanism, Fgfr2 IIIb expression was increased in calvarialsutures and the zygomatic joints allowing cells, which usuallyexpress Fgfr2 IIIc, to respond to a broader set of ligands.Similarity between this mouse mutant and the Apert syndromemouse suggests that the Fgfr2 +/S252W mutation isneomorphic as supported by the observed change in ligand-binding specificity (Yu et al., 2000), rather than being a‘simple’ gain-of-function mutation (Anderson et al., 1998).

Mice created by conditional inactivation of Fgfr2 withDermo1cre/+, specifically targeting Cre expression to disruptsignaling in the chondrocyte or osteocyte lineages, gave furtherinsight into the potential mechanism of the Apert mutation (Yuet al., 2003). These mutant mice had a few features, such as adomed-shaped skull with a midline interfrontal sutural defect,that are similar to our Apert syndrome model. They also had adwarfism phenotype with a shortened axial and appendicularskeleton that differed from our mice by a reduced hypertrophicchondrocyte zone, vertebral abnormalities with non-ossifiedgap in the dorsal midline of both cervical and thoracicvertebrae, absence of the spinous processes, and a lack of tarsaljoints because of failure of cavitation of the cartilaginousanlage. They demonstrated that Fgfr2 is essential for theproliferation of osteoprogenitors and for the maintenance of

osteoblast anabolic function, but that it is not required forosteoblast differentiation. The phenotype of their mice isdifferent from our Apert syndrome mice because themechanism of the Fgfr2 +/S252W mutation is not inactivation.Analysis of our mouse model establishes a potential linkbetween abnormal proliferation and differentiation, andpossibly altered cell fate determination of progenitor cells withdevelopmental abnormalities in Apert syndrome. Abnormalchondrogenesis may play a role in the pathogenesis ofcraniosynostosis syndromes. The Apert mouse model providesan in vivo system for future studies of determinants of cell fatefor the chondrocyte or osteoblast lineages, which are importantin the larger context of bone cell biology.

This work was supported by NIH grants DE11441 (E.W.J.),DE13078 (E.W.J. and J.T.R.), F33DE/HD05706 (J.T.R.), HD38384(J.T.R.), HD24605 (J.T.R.), RR00171 (D.L.H.) and HD39952(D.M.O.).

Supplementary material

Supplementary material for this article is available athttp://dev.biologists.org/cgi/content/full/132/15/3537/DC1

ReferencesAnderson, J., Burns, H. D., Enriquez-Harris, P., Wilkie, A. O. and Heath,

J. K. (1998). Apert syndrome mutations in fibroblast growth factor receptor2 exhibit increased affinity for FGF ligand. Hum. Mol. Genet. 7, 1475-1483.

Arman, E., Haffner-Krausz, R., Chen, Y., Heath, J. K. and Lonai, P.(1998). Targeted disruption of fibroblast growth factor (FGF) receptor 2suggests a role for FGF signaling in pregastrulation mammaliandevelopment. Proc. Natl. Acad. Sci. USA 95, 5082-5087.

Arman, E., Haffner-Krausz, R., Gorivodsky, M. and Lonai, P. (1999).Fgfr2 is required for limb outgrowth and lung-branching morphogenesis.Proc. Natl. Acad. Sci. USA 96, 11895-11899.

Celli, G., LaRochelle, W. J., Mackem, S., Sharp, R. and Merlino, G. (1998).Soluble dominant-negative receptor uncovers essential roles for fibroblastgrowth factors in multi-organ induction and patterning. EMBO J. 17, 1642-1655.

Chen, L., Li, D., Li, C., Engel, A. and Deng, C. X. (2003). A Ser250Trpsubstitution in mouse fibroblast growth factor receptor 2 (Fgfr2) results incraniosynostosis. Bone 33, 169-178.

Cohen, M. M., Jr (2000). Apert syndrome. In Craniosynostosis Diagnosis,Evaluation, and Management (eds. M. M. Cohen, Jr and R. E. McLean), pp.316-353. New York: Oxford University Press.

Cohen, M. M., Jr and Kreiborg, S. (1992). New indirect method forestimating the birth prevalence of the Apert syndrome. Int. J. Oral.Maxillofac. Surg. 21, 107-109.

Cohen, M. M., Jr and Kreiborg, S. (1993). Visceral anomalies in the Apertsyndrome. Am. J. Med. Genet. 45, 758-760.

Debiais, F., Lemonnier, J., Hay, E., Delannoy, P., Caverzasio, J. and Marie,P. J. (2001). Fibroblast growth factor-2 (FGF-2) increases N-cadherinexpression through protein kinase C and Src-kinase pathways in humancalvaria osteoblasts. J. Cell Biochem. 81, 68-81.

De Moerlooze, L., Spencer-Dene, B., Revest, J., Hajihosseini, M., Rosewell,I. and Dickson, C. (2000). An important role for the IIIb isoform offibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelialsignalling during mouse organogenesis. Development 127, 483-492.

Elisseeff, J., Kim, T. K., Ruffner, M. and Williams, C. G. (2005). Cellularphotoencapsulation in hydrogels. In Culture of Cells for Tissue Engineering(eds. I. Freshney and G. Vunjak-Novakovic). Hoboken, NJ: John Wiley andSons (in press).

Eswarakumar, V. P., Monsonego-Ornan, E., Pines, M., Antonopoulou, I.,Morriss-Kay, G. M. and Lonai, P. (2002). The IIIc alternative of Fgfr2 isa positive regulator of bone formation. Development 129, 3783-3793.

Hajihosseini, M. K., Wilson, S., De Moerlooze, L. and Dickson, C. (2001).A splicing switch and gain-of-function mutation in Fgfr2-IIIc hemizygotescauses Apert/Pfeiffer-syndrome-like phenotypes. Proc. Natl. Acad. Sci. USA98, 3855-3860.

Ibrahimi, O. A., Eliseenkova, A. V., Plotnikov, A. N., Yu, K., Ornitz, D. M.

Dev

elop

men

t

Page 12: Abnormalities in cartilage and bone development in the Apert · Bones are formed by one of two types of ossification processes, intramembranous or endochondral bone formation. The

3548

and Mohammadi, M. (2001). Structural basis for fibroblast growth factorreceptor 2 activation in Apert syndrome. Proc. Natl. Acad. Sci. USA 98,7182-7187.

Iseki, S., Wilkie, A. O. and Morriss-Kay, G. M. (1999). Fgfr1 and Fgfr2have distinct differentiation- and proliferation-related roles in the developingmouse skull vault. Development 126, 5611-5620.

Jiang, X., Iseki, S., Maxson, R. E., Sucov, H. M. and Morriss-Kay, G. M.(2002). Tissue origins and interactions in the mammalian skull vault. Dev.Biol. 241, 106-116.

Kouhara, H., Hadari, Y. R., Spivak-Kroizman, T., Schilling, J., Bar-Sagi,D., Lax, I. and Schlessinger J. (1997). A lipid-anchored Grb2-bindingprotein that links FGF-receptor activation to the Ras/MAPK signalingpathway. Cell 89, 693-702.

Kreiborg, S. and Cohen, M. M., Jr (1990). Characteristics of the infant Apertskull and its subsequent development. J. Craniofac. Genet. Dev. Biol. 10,399-410.

Kreiborg, S., Marsh, J. L., Cohen, M. M., Jr, Liversage, M., Pedersen, H.,Skovby, F., Borgesen, S. E. and Vannier, M. W. (1993). Comparativethree-dimensional analysis of CT-scans of the calvaria and cranial base inApert and Crouzon syndromes. J. Craniomaxillofac. Surg. 21, 181-188.

Kreiborg, S., Aduss, H. and Cohen, M. M., Jr (1999). Cephalometric studyof the Apert syndrome in adolescence and adulthood. J. Craniofac. Genet.Dev. Biol. 19, 1-11.

Lemonnier, J., Delannoy, P., Hott, M., Lomri, A., Modrowski, D. andMarie, P. J. (2000). The Ser252Trp fibroblast growth factor receptor-2(FGFR-2) mutation induces PKC-independent downregulation of FGFR-2associated with premature calvaria osteoblast differentiation. Exp. Cell. Res.256, 158-167.

Lemonnier, J., Hay, E., Delannoy, P., Fromigue, O., Lomri, A., Modrowski,D. and Marie, P. J. (2001). Increased osteoblast apoptosis in Apertcraniosynostosis: role of protein kinase C and interleukin-1. Am. J. Pathol.158, 1833-1842.

Le Douarin, N. M., Ziller, C. and Couly, G. F. (1993). Patterning of neuralcrest derivatives in the avian embryo: in vivo and in vitro studies. Dev. Biol.159, 24-49.

Liu, Y. H., Tang, Z., Kundu, R. K., Wu, L., Luo, W., Zhu, D., Sangiorgi,F., Snead, M. L. and Maxson, R. E. (1999). Msx2 gene dosage influencesthe number of proliferative osteogenic cells in growth centers of thedeveloping murine skull: a possible mechanism for MSX2-mediatedcraniosynostosis in humans. Dev. Biol. 205, 260-274.

Lomri, A., Lemonnier, J., Hott, M., de Parseval, N., Lajeunie, E.,Munnich, A., Renier, D. and Marie, P. J. (1998). Increased calvaria celldifferentiation and bone matrix formation induced by fibroblast growthfactor receptor 2 mutations in Apert syndrome. J. Clin. Invest. 101, 1310-1317.

Mansukhani, A., Bellosta, P., Sahni, M. and Basilico, C. (2000). Signalingby fibroblast growth factors (FGF) and fibroblast growth factor receptor 2(FGFR2)-activating mutations blocks mineralization and induces apoptosisin osteoblasts. J. Cell Biol. 149, 1297-1308.

Marie, P. J., Debiais, F. and Hay, E. (2002). Regulation of human cranialosteoblast phenotype by FGF-2, FGFR-2 and BMP-2 signaling. Histol.Histopathol. 17, 877-885.

McLeod, M. J. (1980). Differential staining of cartilage and bone in wholemouse fetuses by alcian blue and alizarin red S. Teratology 22, 299-301.

Miki, T., Bottaro, D. P., Fleming, T. P., Smith, C. L., Burgess, W. H., Chan,A. M. and Aaronson, S. A. (1992). Determination of ligand-bindingspecificity by alternative splicing: two distinct growth factor receptorsencoded by a single gene. Proc. Natl. Acad. Sci. USA 89, 246-250.

Noden, D. M. (1983). The role of the neural crest in patterning of avian cranialskeletal, connective, and muscle tissues. Dev. Biol. 96, 144-165.

Oldridge, M., Zackai, E. H., McDonald-McGinn, D. M., Iseki, S., Morriss-Kay, G. M., Twigg, S. R., Johnson, D., Wall, S. A., Jiang, W., Theda, C.et al. (1999). De novo Alu-element insertions in FGFR2 identify a distinctpathological basis for Apert syndrome. Am. J. Hum. Genet. 64, 446-461.

Opperman, L. A. (2000). Cranial sutures as intramembranous bone growthsites. Dev. Dyn. 219, 472-485.

Ornitz, D. M. and Itoh, N. (2001). Fibroblast growth factors. Genome Biol.2 reviews3005.

Ornitz, D. M. and Marie, P. J. (2002). FGF signaling pathways inendochondral and intramembranous bone development and human geneticdisease. Genes Dev. 16, 1446-1465.

Orr-Urtreger, A., Bedford, M. T., Burakova, T., Arman, E., Zimmer, Y.,Yayon, A., Givol, D. and Lonai, P. (1993). Developmental localization ofthe splicing alternatives of fibroblast growth factor receptor-2 (FGFR2). Dev.

Biol. 158, 475-486.Park, W. J., Theda, C., Maestri, N. E., Meyers, G. A., Fryburg, J. S.,

Dufresne, C., Cohen, M. M., Jr and Jabs, E. W. (1995). Analysis ofphenotypic features and FGFR2 mutations in Apert syndrome. Am. J. Hum.Genet. 57, 321-328.

Petiot, A., Ferretti, P., Copp, A. J. and Chan, C. T. (2002). Induction ofchondrogenesis in neural crest cells by mutant fibroblast growth factorreceptors. Dev. Dyn. 224, 210-221.

Pittenger, M. F., Mackay, A. M., Beck, S. C., Jaiswal, R. K., Douglas, R.,Mosca, J. D., Moorman, M. A., Simonetti, D. W., Craig, S. and Marshak,D. R. (1999). Multilineage potential of adult human mesenchymal stemcells. Science 284, 143-147.

Rice, D. P., Aberg, T., Chan, Y., Tang, Z., Kettunen, P. J., Pakarinen, L.,Maxson, R. E. and Thesleff, I. (2000). Integration of FGF and TWIST incalvarial bone and suture development. Development 127, 1845-1855.

Richtsmeier, J. T., Baxter, L. L. and Reeves, R. H. (2000). Parallels ofcraniofacial maldevelopment in Down syndrome and Ts65Dn mice. Dev.Dyn. 217, 137-145.

Selleck, M. A., Scherson, T. Y. and Bronner-Fraser, M. (1993). Origins ofneural crest cell diversity. Dev. Biol. 159, 1-11.

Slaney, S. F., Oldridge, M., Hurst, J. A., Moriss-Kay, G. M., Hall, C. M.,Poole, M. D. and Wilkie, A. O. (1996). Differential effects of FGFR2mutations on syndactyly and cleft palate in Apert syndrome. Am. J. Hum.Genet. 58, 923-932.

von Gernet, S., Golla, A., Ehrenfels, Y., Schuffenhauer, S. and Fairley, J.D. (2000). Genotype-phenotype analysis in Apert syndrome suggestsopposite effects of the two recurrent mutations on syndactyly and outcomeof craniofacial surgery. Clin. Genet. 57, 137-139.

Wilkie, A. O., Slaney, S. F., Oldridge, M., Poole, M. D., Ashworth, G. J.,Hockley, A. D., Hayward, R. D., David, D. J., Pulleyn, L. J., Rutland, P.et al. (1995). Apert syndrome results from localized mutations of FGFR2and is allelic with Crouzon syndrome. Nat. Genet. 9, 165-172.

Wilkinson, D. G. (1992). In Situ Hybridization: A Practical Approach.London, UK: Oxford University Press.

Xu, X., Weinstein, M., Li, C., Naski, M., Cohen, R. I., Ornitz, D. M., Leder,P. and Deng, C. (1998). Fibroblast growth factor receptor 2 (FGFR2)-mediated reciprocal regulation loop between FGF8 and FGF10 is essentialfor limb induction. Development 125, 753-765.

Yu, K., Herr, A. B., Waksman, G. and Ornitz, D. M. (2000). Loss offibroblast growth factor receptor 2 ligand-binding specificity in Apertsyndrome. Proc. Natl. Acad. Sci. USA 97, 14536-14541.

Yu, K., Xu, J., Liu, Z., Sosic, D., Shao, J., Olson, E. N., Towler, D. A. andOrnitz, D. M. (2003). Conditional inactivation of FGF receptor 2 revealsan essential role for FGF signaling in the regulation of osteoblast functionand bone growth. Development 130, 3063-3074.

Development 132 (15) Research article

Dev

elop

men

t