14
Developmental Cell, Vol. 1, 277–290, August, 2001, Copyright 2001 by Cell Press The Transcription Factors L-Sox5 and Sox6 Are Essential for Cartilage Formation proliferate and abundantly produce the cartilage extra- cellular matrix. Chondroblasts then establish cartilage growth plates, in which they proceed layer by layer Patrick Smits, Ping Li, Jennifer Mandel, Zhaoping Zhang, Jian Ming Deng, Richard R. Behringer, through further steps of differentiation. They proliferate Benoit de Crombrugghe, unidirectionally in characteristic cell columns, undergo and Ve ´ ronique Lefebvre 1,2 prehypertrophy and growth arrest, and finally hypertro- Department of Molecular Genetics phy and apoptosis. The University of Texas M.D. Anderson Chondrogenesis is controlled by a complex interplay Cancer Center of regulatory factors (Cancedda et al., 1995; Karsenty, Houston, Texas 77030 1998 and 2001; DeLise et al., 2000). Secreted factors include TGF- and bone morphogenetic (BMP) proteins, the parathyroid hormone-related peptide (PTHrP), and Summary Sonic and Indian Hedgehog (Shh and Ihh). Whereas nu- merous patterning transcription factors have been iden- L-Sox5 and Sox6 are highly identical Sry-related tran- tified, including homeobox, Hox, Pax, forkhead, and he- scription factors coexpressed in cartilage. Whereas lix-loop-helix factors, only two transcription factors, Sox5 and Sox6 single null mice are born with mild Sox9 and Cbfa1, have been demonstrated to control the skeletal abnormalities, Sox5; Sox6 double null fetuses differentiation per se of chondrocytes. Sox9, an Sry- die with a severe, generalized chondrodysplasia. In related HMG box transcription factor (Wegner, 1999), is these double mutants, chondroblasts poorly differen- expressed from the prechondrocytic to the prehypertro- tiate. They express the genes for all essential cartilage phic chondrocyte stages (Ng et al., 1997; Zhao et al., extracellular matrix components at low or undetect- 1997). Experiments in mouse chimeras and teratomas able levels and initiate proliferation after a long delay. using Sox9 / embryonic stem (ES) cells have demon- All cartilages are thus extracellular matrix deficient strated that it is required for prechondrocyte aggrega- and remain rudimentary. While chondroblasts in the tion and activation of cartilage early markers, including center of cartilages ultimately activate prehypertro- the Col2a1 collagen type 2 gene (Bi et al., 1999). Consis- phic chondrocyte markers, epiphyseal chondroblasts tent with these data, ectopic expression of SOX9 in ectopically activate hypertrophic chondrocyte mark- transgenic mice was shown to result in Col2a1 activation ers. Thick intramembranous bone collars develop, but (Bell et al., 1997). SOX9 may directly activate cartilage- the formation of cartilage growth plates and endo- specific genes, since it binds and activates cartilage- chondral bones is disrupted. L-Sox5 and Sox6 are thus specific enhancers in Col2a1 (Lefebvre et al., 1997; Bell redundant, potent enhancers of chondroblast func- et al., 1997), Col11a2 (Bridgewater et al., 1998), and CD- tions, thereby essential for endochondral skeleton for- RAP (Xie et al., 1999). Cbfa1, a runt family transcription mation. factor, is expressed early in osteo-chondroprogenitor cells and maintained throughout osteoblast differentia- Introduction tion (Ducy et al., 1997). It is turned off in prechondrocytes and chondroblasts but reactivated in prehypertrophic Chondrogenesis is an essential process in vertebrate and hypertrophic chondrocytes. Cbfa1 null mice have skeleton development. It leads to the formation of man- proven that it is required for osteoblast differentiation datory cartilage scaffolds, upon which axial, appendicu- in all bones and hypertrophic chondrocyte differentia- lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process called endochondral ossification. A single cell 1997). Forced expression of Cbfa1 in chondroblasts in type, the chondrocyte, is present in cartilage, where it transgenic mice has confirmed the ability of Cbfa1 to follows a multistep differentiation program to ensure induce chondrocyte hypertrophy (Takeda et al., 2001). successively cartilage formation, growth, and preossifi- We previously showed that the transcription factors cation remodeling (Cancedda et al., 1995). Chondro- L-Sox5 (a long product of the Sox5 gene) and Sox6 are progenitor cells emerge during embryogenesis from coexpressed with Sox9 in all cartilages (Lefebvre et al., the cranial neural crest, sclerotome, and lateral plate 1998). They share 67% identity with each other, with mesoderm. After migration to the various sites of chon- more than 90% in the HMG box DNA binding domain drogenesis, they proliferate and differentiate into pre- and a coiled-coil dimerization domain, but share with chondrocytes, which aggregate into precartilaginous Sox9 only 50% identity in the HMG box domain and no condensations, growth-arrest, and activate cartilage significant identity outside this domain. Sox9 harbors a transactivation domain, but L-Sox5 and Sox6 do not. early marker genes. Differentiation into true cartilage L-Sox5 and Sox6 homo- and heterodimerize and prefer- tissue-forming cells follows. At this stage, the cells are entially bind pairs of DNA recognition sites, whereas best referred to as chondroblasts, since they actively Sox9 binds single DNA sites as a monomer. The three Sox proteins bind in vitro to several sites in a Col2a1 1 Correspondence: [email protected] cartilage-specific enhancer and cooperate to activate 2 Present address: Department of Biomedical Engineering, Lerner this enhancer and the Col2a1 and aggrecan genes in Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195. cultured cells (Lefebvre et al., 1998). They also bind

Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell, Vol. 1, 277–290, August, 2001, Copyright 2001 by Cell Press

The Transcription Factors L-Sox5 and Sox6Are Essential for Cartilage Formation

proliferate and abundantly produce the cartilage extra-cellular matrix. Chondroblasts then establish cartilagegrowth plates, in which they proceed layer by layer

Patrick Smits, Ping Li, Jennifer Mandel,Zhaoping Zhang, Jian Ming Deng,Richard R. Behringer,

through further steps of differentiation. They proliferateBenoit de Crombrugghe,unidirectionally in characteristic cell columns, undergoand Veronique Lefebvre1,2

prehypertrophy and growth arrest, and finally hypertro-Department of Molecular Geneticsphy and apoptosis.The University of Texas M.D. Anderson

Chondrogenesis is controlled by a complex interplayCancer Centerof regulatory factors (Cancedda et al., 1995; Karsenty,Houston, Texas 770301998 and 2001; DeLise et al., 2000). Secreted factorsinclude TGF-� and bone morphogenetic (BMP) proteins,the parathyroid hormone-related peptide (PTHrP), andSummarySonic and Indian Hedgehog (Shh and Ihh). Whereas nu-merous patterning transcription factors have been iden-L-Sox5 and Sox6 are highly identical Sry-related tran-tified, including homeobox, Hox, Pax, forkhead, and he-scription factors coexpressed in cartilage. Whereaslix-loop-helix factors, only two transcription factors,Sox5 and Sox6 single null mice are born with mildSox9 and Cbfa1, have been demonstrated to control theskeletal abnormalities, Sox5; Sox6 double null fetusesdifferentiation per se of chondrocytes. Sox9, an Sry-die with a severe, generalized chondrodysplasia. Inrelated HMG box transcription factor (Wegner, 1999), isthese double mutants, chondroblasts poorly differen-expressed from the prechondrocytic to the prehypertro-tiate. They express the genes for all essential cartilagephic chondrocyte stages (Ng et al., 1997; Zhao et al.,extracellular matrix components at low or undetect-1997). Experiments in mouse chimeras and teratomasable levels and initiate proliferation after a long delay.using Sox9�/� embryonic stem (ES) cells have demon-All cartilages are thus extracellular matrix deficientstrated that it is required for prechondrocyte aggrega-and remain rudimentary. While chondroblasts in thetion and activation of cartilage early markers, includingcenter of cartilages ultimately activate prehypertro-the Col2a1 collagen type 2 gene (Bi et al., 1999). Consis-phic chondrocyte markers, epiphyseal chondroblaststent with these data, ectopic expression of SOX9 inectopically activate hypertrophic chondrocyte mark-transgenic mice was shown to result in Col2a1 activationers. Thick intramembranous bone collars develop, but(Bell et al., 1997). SOX9 may directly activate cartilage-the formation of cartilage growth plates and endo-specific genes, since it binds and activates cartilage-chondral bones is disrupted. L-Sox5 and Sox6 are thusspecific enhancers in Col2a1 (Lefebvre et al., 1997; Bellredundant, potent enhancers of chondroblast func-et al., 1997), Col11a2 (Bridgewater et al., 1998), and CD-tions, thereby essential for endochondral skeleton for-RAP (Xie et al., 1999). Cbfa1, a runt family transcription

mation.factor, is expressed early in osteo-chondroprogenitorcells and maintained throughout osteoblast differentia-

Introduction tion (Ducy et al., 1997). It is turned off in prechondrocytesand chondroblasts but reactivated in prehypertrophic

Chondrogenesis is an essential process in vertebrate and hypertrophic chondrocytes. Cbfa1 null mice haveskeleton development. It leads to the formation of man- proven that it is required for osteoblast differentiationdatory cartilage scaffolds, upon which axial, appendicu- in all bones and hypertrophic chondrocyte differentia-lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al.,a process called endochondral ossification. A single cell 1997). Forced expression of Cbfa1 in chondroblasts intype, the chondrocyte, is present in cartilage, where it transgenic mice has confirmed the ability of Cbfa1 tofollows a multistep differentiation program to ensure induce chondrocyte hypertrophy (Takeda et al., 2001).successively cartilage formation, growth, and preossifi- We previously showed that the transcription factorscation remodeling (Cancedda et al., 1995). Chondro- L-Sox5 (a long product of the Sox5 gene) and Sox6 areprogenitor cells emerge during embryogenesis from coexpressed with Sox9 in all cartilages (Lefebvre et al.,the cranial neural crest, sclerotome, and lateral plate 1998). They share 67% identity with each other, withmesoderm. After migration to the various sites of chon- more than 90% in the HMG box DNA binding domaindrogenesis, they proliferate and differentiate into pre- and a coiled-coil dimerization domain, but share withchondrocytes, which aggregate into precartilaginous Sox9 only 50% identity in the HMG box domain and nocondensations, growth-arrest, and activate cartilage significant identity outside this domain. Sox9 harbors a

transactivation domain, but L-Sox5 and Sox6 do not.early marker genes. Differentiation into true cartilageL-Sox5 and Sox6 homo- and heterodimerize and prefer-tissue-forming cells follows. At this stage, the cells areentially bind pairs of DNA recognition sites, whereasbest referred to as chondroblasts, since they activelySox9 binds single DNA sites as a monomer. The threeSox proteins bind in vitro to several sites in a Col2a11 Correspondence: [email protected] enhancer and cooperate to activate2 Present address: Department of Biomedical Engineering, Lernerthis enhancer and the Col2a1 and aggrecan genes inResearch Institute, Cleveland Clinic Foundation, Cleveland, Ohio

44195. cultured cells (Lefebvre et al., 1998). They also bind

Page 2: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell278

Page 3: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

L-Sox5 and Sox6 Control Chondrogenesis279

critical sequences in two cartilage-specific enhancers of Limited Skeletal Abnormalities in Sox5�/�

and Sox6�/� Single Null Mutantsthe Col11a2 gene (Bridgewater et al., 1998). Collectively,Mice with up to three mutant alleles of Sox5 and Sox6these findings strongly suggested that L-Sox5 and Sox6were born alive with the expected Mendelian ratio,could contribute to the activation of the chondrocyteweight, and size. Single heterozygotes were indistin-program, acting as redundant factors in cooperationguishable from wild-type littermates on to adulthood.with Sox9 but with different roles than Sox9. To test thisDouble heterozygotes were slightly smaller and leanerhypothesis, we created null mutations of Sox5 and Sox6postnatally, but viable and fertile. In skeletal prepara-in the mouse. We show that Sox5�/� and Sox6�/� singletions, in which cartilage was stained with alcian bluenull mice were born with few skeletal defects, but thatand bone with alizarin red, double heterozygotes oftenSox5�/�; Sox6�/� double null embryos developed a se-showed a slightly shorter and irregularly mineralizedvere, generalized chondrodysplasia. Detailed histologysternum but were otherwise normal (data not shown).and gene expression analysis revealed that L-Sox5 and

Sox5�/� mice died at birth with respiratory distress.Sox6 act mostly redundantly to enhance chondroblastThey had a cleft secondary palate and a short chondro-functions, controlling both expression of extracellularcranium (Figures 2A and 2B). Ribs also were short, form-matrix genes and cell proliferation.ing a bell-shaped thoracic cage (Figure 2C). Mineraliza-tion of a few endochondral elements was slightlydelayed, including that of vertebral bodies (Figure 2D),Resultsnasal, and presphenoid bones (data not shown). Thesternum of Sox6�/� mice was short, bent inwards, andGeneration of Sox5; Sox6 Mutant Miceoften presenting ectopically mineralized intersternebraeThe mouse Sox5 and Sox6 genes being split into multiple(Figures 2E and 2F). Most Sox6�/� mice died at birth,small exons spread over more than 100 kb (data notfailing to breathe. Those that survived developed severeshown), we chose to disrupt them by introducing a lacZ/dwarfism within a week, stopped feeding by the end ofneo cassette into a coding exon preceding the dimeriza-the second week, and died soon afterwards (data nottion and DNA binding motif sequences (Figure 1A). Anshown). Thus, the Sox5 and Sox6 single null mutationsinternal ribosome entry site (IRES) was placed upstreamwere both early lethal, but each affected only the sizeof the lacZ gene, allowing expression of �-galactosidaseand mineralization rate of a small, distinct subset ofwith a Sox5- or Sox6-specific pattern. ES cell clonesendochondral elements.with a Sox5 or Sox6 heterozygous mutation (Figure 1B)

were obtained, from which mouse chimeras and, subse-quently, mice with all combinations of Sox5 and Sox6 Severe and Generalized Chondrodysplasiamutant alleles were derived. in Sox5�/�; Sox6�/� Double Null Embryos

The null mutations were confirmed by Northern analy- Up to E15.5, Sox5�/�; Sox6�/� embryos were recoveredsis. RNA from total embryos was hybridized with probes at the expected 1:16 ratio from crosses of double hetero-recognizing the L-Sox5 and Sox6 transcripts 3� of the zygotes. From E13.5, they featured a rounded head,mutations (Figure 1C). For each gene inactivation, the short snout, trunk, limbs, and tail, and a prominent abdo-hybridization signals were, as expected, half as intense men (Figure 3A). They died around E16.5 from heartin heterozygous as in wild-type samples, and no signal failure, as indicated by generalized edema and accumu-was detected in RNA from null homozygotes. lation of blood in the liver, posthepatic segment of the

Staining of Sox5�/� and Sox6�/� single null embryos inferior vena cava, and right atrium of the heart (Figureswith X-gal revealed lacZ expression in every cartilage 3B and 3C). The short vertebral column and incomplete(Figure 1D), confirming previous Sox5 and Sox6 expres- thoracic cage (see below) caused anteroposterior com-sion data (Lefebvre et al., 1998). Staining was generally pression of otherwise normal internal organs, ballooningmore intense in Sox5�/� than in Sox6�/� embryos, possi- of the abdomen, and thus likely, the ultimate heart fail-bly reflecting differences in gene expression levels. ure. In one exception, a Sox5�/�; Sox6�/� mouse wasStaining in the telencephalon, corresponding to expres- recovered at birth (Figure 3D). Internal organs were ex-sion of Sox5 and Sox6 in the neopallial cortex (Lefebvre truded, but there was no sign of bleeding, indicatinget al., 1998), was also more intense in Sox5�/� than in that the body wall had ruptured in utero and not upon

delivery. This extrusion of internal organs likely releasedSox6�/� embryos.

Figure 1. Inactivation of Sox5 and Sox6(A) Schematic representation of alleles, targeting vectors, probes, and primers. The exon targeted in Sox5 and Sox6 is shown as an openbox, and the segments of flanking introns included in the targeting vectors are shown as thick lines. The lacZ/neo and tk cassettes andSouthern probes are indicated. Forward and reverse primers used for PCR genotyping are shown as arrowheads pointing down and up,respectively. DNA fragments revealed in Southern analysis are indicated as arrows and lines, with the restriction enzymes used to generatethem, their size, and the probes that identified them. A, Asp718; B, BamHI; Bg, BgIII; H, HinDIII.(B) Southern analysis of ES cell clones. Genomic DNA from wild-type (�/�) and heterozygous (�/�) clones for each gene knockout (KO) wasdigested with restriction enzymes and hybridized with DNA probes as indicated. Bands corresponding to the wild-type and mutant allelesare identified by their size.(C) Northern blot analysis of RNA from E12.5 littermates generated by crossing Sox5 or Sox6 single heterozygotes. Blots were hybridizedwith L-Sox5 and Sox6 RNA probes and rehybridized with a 28S RNA probe as a control for RNA loading.(D) X-gal-stained Sox5�/� and Sox6�/� E15.5 embryos. LacZ was expressed in all cartilages and in the telencephalon in both types of embryos.

Page 4: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell280

Figure 2. Skeletal Abnormalities of Sox5 and Sox6 Single Null Mutants at Birth

(A) Sox5�/� mice had a cleft secondary palate. In this and all following panels, wild-type is on the left and homozygous null on the right.(B) Ventral view of the chondrocranium and upper jaw. This and all following panels are pictures of skeletal preparations. The maxillary (MS)and palatal shelves (PS) were elevated to a horizontal position and almost fused in the midline in the wild-type mouse. In the mutant mouse,they were still vertical. The mutant pterigoid processes (PP) did not move toward the midline. The mutant chondrocranium was shorter overall(double arrow).(C) The ribs of Sox5�/� mice were short and formed a narrow, bell-shaped thoracic cage. In most mutant mice, as shown here, the seventhpair of ribs did not attach to the sternum (R7; arrow).(D) Mineralization of cervical vertebral bodies (VB; arrow) was delayed in Sox5�/� mice.(E) The sternum of Sox6�/� mice was short and bent inwards.(F) Mineralization of sternebrae (ST) spread into intersternebrae (IS) in Sox6�/� mice.

internal organ pressure, allowing this mouse to live until were unable to develop cartilages and produce a typicalalcian blue-stainable extracellular matrix.birth and thus supporting the notion that the chondro-

dysplasia of double null embryos indirectly caused their The skeleton of the double null newborn mouse alsofailed to stain significantly with alcian blue (Figures 3G–heart failure.

While X-gal staining of Sox5�/�; Sox6�/� embryos was 3I). Staining with alizarin red was seen in all craniofacialbones and clavicles that form through a cartilage-inde-strong in all expected sites (Figure 3E), staining with

alcian blue was barely detectable (Figure 3F). Both stain- pendent intramembranous mode of ossification (Figures3G and 3I). It was also seen in the basioccipital element,ings confirmed underdevelopment of the skeleton. Thus,

while cells expressing the Sox-lacZ genes and thus com- neural arches of thoracic vertebrae, rib dorsal segments,and humeri and femurs, which are the first cartilages tomitted to chondrogenesis were present in all sites, they

Page 5: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

L-Sox5 and Sox6 Control Chondrogenesis281

mineralize and undergo endochondral ossification in the lightly staining with alcian blue (Figure 5F). Interestingly,in thoracic vertebral bodies wild-type chondrocytesembryo. However, the mineralized humerus segment

was hollow, suggesting that it corresponded to an intra- were fully hypertrophic and double null cells were alsosomewhat enlarged, suggesting an attempt to undergomembranous bone collar rather than mineralized carti-

lage or endochondral bone (Figure 3H). hypertrophy (Figure 5G). The same attempt was seen inthe Sox5�/�; Sox6�/� humerus and further suggested byThus, in contrast to single null mice, Sox5; Sox6 dou-

ble null embryos developed a very severe and general- the presence of an intramembranous bone collar, whoseinduction requires that adjacent chondrocytes have be-ized chondrodysplasia, characterized by a virtual ab-

sence of bona fide cartilages. They proved that Sox5 come prehypertrophic (Figure 5H). This bone collar wasabnormally thick and extended far around the epiphy-and Sox6 genetically interact to fulfill essential functions

in chondrogenesis. ses, and there were no growth plates and no endochon-dral bone. The double null basioccipital element, themost advanced skeletal element, also featured intra-Normal Differentiation of Sox5�/�; Sox6�/�

membranous bone in surface. Adjacent chondrocytesPrechondrocyteswere enlarged and mineralizing the cartilage matrix,Precartilaginous condensations were seen in all ex-proving their reaching terminal differentiation (Figurespected sites in E12.5 double null embryos (Figure 4A).5I and 5J).In sites of less advanced chondrogenesis, for example,

Thus, Sox5�/�; Sox6�/� chondrocytes never producedhindlimb buds, wild-type and mutant condensationsa typical cartilage extracellular matrix and never formedwere similar in size and cell numbers. Cells were pre-growth plates, but nevertheless ultimately acquired pre-chondrocytic, that is, small and tightly packed. In morehypertrophic and hypertrophic cell properties.advanced sites, scapulas, for example, where wild-type

cells were becoming chondroblastic—that is, accumu-lating alcian blue-stainable matrix—mutant cells were No Overt Differentiation of Sox5�/�; Sox6�/�

still prechondrocytic. The mesenchymal marker colla- Chondroblastsgen type 1 gene (Col1a1) was inactive and Sox9 RNA To actively assemble the cartilage extracellular matrix,levels were similar in all wild-type and double null pre- chondroblasts upregulate early markers upon differenti-cartilages (Figure 4B). Interestingly, Col2a1, an early car- ation from prechondrocytes and activate stage-specifictilage matrix marker upregulated upon chondroblast dif- markers. In E14.5–E16.5 double null cartilages, the ex-ferentiation, was expressed in double null precartilage, pression levels of all major early cartilage matrix compo-but at a lower level than in wild-type precartilage. By nents, i.e., collagen type 2, aggrecan, and link protein,E12.5, Sox5�/�; Sox6�/� cells had thus turned off their remained much lower than with wild-type elements (Fig-mesenchymal phenotype and become prechondro- ure 6A). This was also the case in minor early compo-cytic, but were failing to rapidly differentiate into chon- nents, such as the collagen types 9 (Col9a2) and 11droblasts. (Col11a2) (data not shown). The chondroblast-specific

At E12.5, the most distal condensations of wild-type COMP and matrilin-1 genes remained virtually inactivehindlimb buds are still accumulating new prechondro- in double null cartilages (Figure 6A). Very low expressioncytes in periphery, while core cells are preparing them- levels were detectable only in the most advanced carti-selves to differentiate into chondroblasts. Whereas Sox9 lages at E15.5–16.5 (data not shown). The same wasexpression was high throughout these condensations, seen for other chondroblast markers, including theSox5 and Sox6 expression was only detectable at low genes for PG-Lb/epiphycan, perlecan, and matrix Glalevels in core cells (Figure 4C). In the second to most protein (data not shown).distal condensations, where chondroblasts were overtly The genes for the BMP Gdf5 and for fibromodulin aredifferentiating, the hybridization signals for the three Sox expressed early in prechondrocytes and later in peri-genes were similarly high and evenly distributed. The chondrial cells but not in chondroblasts. At E14.5, bothexpression pattern of Col2a1 was similar to that of Sox5 Gdf5 and fibromodulin RNAs were found in wild-typeand Sox6. These data are consistent with the notion that perichondriums and Sox5�/�; Sox6�/� cartilages (FigureSox9 is needed for prechondrocyte differentiation, and 6B). This result thus confirms that mutant cartilage cellsSox5 and Sox6, as well as Sox9, are needed for chon- were still prechondrocytic and, because Sox5 and Sox6droblast differentiation. are not expressed in the perichondrium, supports the

notion that the maturation of perichondrial cells is linkedto that of chondroblasts.Severe Underdevelopment and Disorganization

of Sox5�/�; Sox6�/� Cartilages Cell proliferation in developing cartilages is best illus-trated by staining for the proliferative cell nuclear anti-By E16.5, whereas Sox5�/�; Sox6�/� intramembranous

craniofacial bones and teeth were histologically normal, gen (PCNA) present in cells at the S, G2, and M phasesof the cell cycle and absent in cells at the G1 and G0Sox5�/�; Sox6�/� cartilages were still underdeveloped

and showing poor or no staining with alcian blue (Figures phases. For example, PCNA was undetectable in pre-chondrocytes in the ventral segments of E12.5 wild-5A–5D). More specifically, in digits, where wild-type

chondrocytes were forming growth plates, Sox5�/�; type ribs but detectable in chondroblasts in the moreadvanced dorsal segments (Figure 6C). In E16.5 wild-Sox6�/� cells were still prechondrocytic (Figure 5E).

Whereas wild-type tibias were featuring growth plates type humerus growth plates, most chondroblasts in thereserve and columnar zones stained intensely for PCNA,and endochondral bone, mutant tibias were only show-

ing first signs of chondroblast differentiation in the core whereas prehypertrophic and hypertrophic chondro-cytes did not (Figure 6D). In E16.5 Sox5�/�; Sox6�/�region, with accumulation of some extracellular matrix

Page 6: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell282

Figure 3. Severe, Generalized Chondrodysplasia in Sox5�/�; Sox6�/� Embryos

(A) Sox5�/� (left) and Sox5�/�; Sox6�/� (right) embryos at E16.5. The double null embryo had a small, rounded head, short snout with thetongue sticking out, short limbs, trunk, and tail, and a prominent abdomen.(B) Double null embryo found dead at E16.5, with generalized edema and a congested abdomen.(C) Hematoxylin and eosin staining of midsagittal sections of a Sox6�/� E16.5 embryo (left) and a dead Sox5�/�; Sox6�/� littermate (right). Thedouble null embryo displayed numerous skeletal defects, including severe hypoplasia of the vertebral column (C1, T1, L1, and S1 indicatethe first cervical, thoracic, lumbar, and sacral vertebrae, respectively). The liver, posthepatic segment of the inferior vena cava (arrow), andright atrium of the heart were dilated with blood.(D) Double null mouse born with abdominal organs extruded outside the body cavity. In the back view, the skin was torn apart to show theprotruded spinal cord in the thoracolumbar region.(E) X-gal staining of Sox5�/�; Sox6�/� (left) and Sox5�/�; Sox6�/� (right) littermates at E15.5. Staining was strong in all sites of chondrogenesisand in the telencephalon in both embryos.

Page 7: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

L-Sox5 and Sox6 Control Chondrogenesis283

embryos, PCNA staining was still negative in all least and hypertrophic chondrocytes but not in chon-droblasts, was expressed at high levels throughout theadvanced sites of chondrogenesis, such as digital rays

(Figure 6E). In more advanced sites, such as Meckel’s entire mutant cartilage. Similar results were obtainedfor VEGF, another marker of hypertrophic chondrocytescartilage, positive staining for PCNA was seen in most

cells at levels close to those seen in wild-type early (data not shown). Thus, unable to express a fully differ-entiated chondroblastic phenotype, Sox5�/�; Sox6�/�differentiating chondroblasts (Figure 6F). In the hu-

merus, groups of cells positive for PCNA were seen only cartilage cells activated a number of prehypertrophicand hypertrophic markers, but in a disrupted temporalin the lateral sides of epiphyseal cartilages (Figure 6G).

Most cells in the core region were negative or weakly and spatial order, preventing establishment of cartilagegrowth plates.positive. These data further suggest that most cells in

the epiphyses had precociously reached a postmitotic In the bone collars of mutant humeri, differentiatedosteoblasts were expressing PPR, Ptc1, and Cbfa1 (Fig-prehypertrophic or hypertrophic differentiation stage.

Thus, Sox5�/�; Sox6�/� chondroblasts never assumed ure 7) and depositing a von Kossa-positive mineralizedbone matrix (data not shown). Osteoclasts/chondro-a fully active phenotype. Most, if not all, cartilage matrix

genes remained severely downregulated. Cell prolifera- clasts were expressing MMP9 (Figure 7) and tartrate-resistant acid phosphatase (data not shown). Red bloodtion was resumed after a long prechondrocytic block,

but then maintained only for a short period of time, as cells were recognized at their discoid morphology (datanot shown). Thus, bone had the potential to developthe cells appeared to mature rapidly toward late chon-

drocyte differentiation stages. normally in double null embryos.

DiscussionDisorganized Maturation of Sox5�/�; Sox6�/�

ChondrocytesThe maturation defects of Sox5�/�; Sox6�/� chondro- Essential Redundant and Nonredundant Roles

for L-Sox5 and Sox6cytes were further analyzed in humeri at E16.5 (Figure7). All mutant cartilage cells still expressed the early By showing that Sox5 and Sox6 single null embryos

developed distinct mild skeletal abnormalities, whereaschondrocyte markers Sox9 (Figure 7) and Col2a1 (datanot shown). FGFR3, which is normally expressed at low Sox5; Sox6 double null embryos featured major and

generalized cartilage defects, this study has demon-levels in prechondrocytes and epiphyseal reserve chon-droblasts and upregulated in columnar chondroblasts, strated that Sox5 and Sox6 play essential roles in chon-

drogenesis and that they are mostly but not entirelywas expressed only at low levels in the mutant humerus,consistent with the absence of columnar chondroblasts. redundant. Sox5�/�; Sox6�/� fetuses died in utero, most

likely as a consequence of their severe skeletal defects.The prehypertrophic markers PPR and Ihh were ex-pressed at close to wild-type levels in mutant chondro- Similarly, severe cases of human osteochondrodysplas-

ias have also been shown to result in stillbirth. For exam-cytes located, as expected, in the core of the humerusand lining the bone collar. Ptc1, a transcriptional target ple, Blomstrand human fetuses, which have mutations

in PPR (Jobert et al., 1998), develop a dwarfism as severeof Ihh signaling, was highly expressed in the whole mu-tant cartilage. In E16.5 wild-type growth plates, Ptc1 as that of Sox5�/�; Sox6�/� mouse mutants and also die

from cardiac failure, with a prominent abdomen andexpression is restricted to chondrocyte layers adjacentto and overlapping the Ihh expression domain, but in generalized edema but no obvious anomaly of other

organs (Leroy et al., 1996). The skeletal defects ofE12.5 cartilages, which have not yet formed growthplates and are much smaller, Ptc1 is expressed in most Sox5�/� and Sox6�/� mice likely resulted from an insuffi-

cient level of expression of the intact gene in the affectedof the cartilage (St-Jacques et al., 1999). Therefore, theexpression domain of Ptc1 in the E16.5 Sox5�/�; Sox6�/� elements to allow this gene to fully compensate for the

mutation of its relative. Differences in X-gal-staining in-humerus was fairly normal, considering its small sizeand lack of growth plates. PTHrP, another target of Ihh tensity between Sox5 and Sox6 single null embryos sup-

port this hypothesis. Indeed, staining in appendicularsignaling, was detectable at normal levels and, as ex-pected, in the periarticular region of mutant cartilages. cartilages was strong in each type of mice and no abnor-

malities were seen there, whereas staining was strongerCol10a1, an abundant marker of prehypertrophic andhypertrophic chondrocytes, was inactive in mutant in the head cartilages of Sox5 null than of Sox6 null

mice, and only Sox5 null mice displayed craniofacialchondrocytes that expressed Ihh and PPR, but was ec-topically expressed at low levels in most epiphyseal defects. The mildness of skeletal defects in single null

mice indicates that L-Sox5 and Sox6 efficiently functioncells. Cbfa1, which is active in wild-type prehypertrophic

(F) Alcian blue staining of E14.5 Sox5�/�; Sox6�/� (left) and Sox5�/�; Sox6�/� (right) littermates. Cartilages were rudimentary and barely stainedin the double null embryo.(G) Skeletal preparation of the Sox5�/�; Sox6�/� newborn mouse shown in (D). No cartilage was detected upon alcian blue staining. Alizarinred staining was positive in the intramembranous bones of the craniofacial skeleton and in humeri (h), femurs (f), and thoracic vertebral archesand dorsal segments of the ribs (v � r).(H) End-on, high-magnification view of a humerus of the Sox5�/�; Sox6�/� newborn mouse. Staining with alizarin red was hollow.(I) Dorsal view of the base of the skull of wild-type (left) and Sox5�/�; Sox6�/� (right) newborn mice. In the wild-type, cartilages strongly stainedwith alcian blue, and intramembranous and endochondral bones stained with alizarin red. In the mutant, positive staining was obtained onlyfor intramembranous bones and the endochondral basioccipital element (bo).

Page 8: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell284

Figure 4. Precartilage Formation in Sox5�/�; Sox6�/� E12.5 Embryos

(A) Alcian blue staining of sagittal sections through hindlimb paws and scapulas of wild-type (left) and Sox5�/�; Sox6�/� (right) littermates.Top, double null digital rays were normal. Middle, high-magnification views of the areas marked H in top panels. Cells were normal in numberand morphology in double null digital rays. Bottom, chondroblasts were differentiating in the wild-type scapula, starting to accumulate alcianblue-positive matrix, whereas all cells were still prechondrocytic in the mutant scapula.(B) Sagittal sections of cervical and thoracic prevertebrae of wild-type (left) and Sox5�/�; Sox6�/� (right) littermates. Top, wild-type prevertebraestained significantly with alcian blue but mutant prevertebrae did not. Other panels show RNA in situ hybridizations of adjacent sections.Col1a1 expression was absent in prevertebral cartilages and intense in surrounding tissues in both embryos. Sox9 expression was similarlyhigh in wild-type and mutant prevertebrae. Col2a1 expression was less intense in double null than in wild-type prevertebrae.(C) Sequence of gene activation in developing cartilages of wild-type hindlimb buds. On the left, alcian blue staining was negative in the mostdistal precartilaginous condensation (1) and progressively stronger in the more developmentally advanced proximal condensations (2 and 3).The box encompasses the area magnified in other panels. The other panels show RNA in situ hybridizations of adjacent sections. Sox9expression was already strong in the entire most distal condensation, whereas Sox5, Sox6, and Col2a1 were only starting to be expressedin the core of this condensation. All four genes were expressed at high levels throughout the more proximal condensations.

independently of each other in vivo. This conclusion is heterozygotes survived, demonstrating essential, uniqueroles for each gene. Sox5�/� mice were born with a cleftconsistent with in vitro evidence that the proteins bind

DNA as homodimers as well as heterodimers (Lefebvre palate and small thoracic cage, which caused respira-tory distress. No other defects were detected upon his-et al., 1998).

Sox5�/� and Sox6�/� mice died in early life, but double tological analysis, indicating a primary role for Sox5 in

Page 9: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Figure 5. Cartilage Defects in E15.5–16.5 Sox5�/�; Sox6�/� Embryos

Alcian blue (A–I) and von Kossa (J) stainings of sagittal (A–F, H–J) and transverse (G) sections of E16.5 (A–F, I–J) and E15.5 (G–H) embryos.In each panel, a Sox5�/�; Sox6�/� embryo is on the left and a Sox5�/�; Sox6�/� littermate on the right.(A) Face region. The cartilage primordiums of the turbinate bones (tu) stained positively with alcian blue in the wild-type but not in the double-mutant embryo. The intramembranous maxillary (mx) and mandibular (mn) bones and the primordiums of molar (m) and incisor (i) teeth werehistologically normal. The secondary palate (p) was severely malformed in the double null embryo.(B) Neck region. All endochondral elements were hypoplastic and failed to stain with alcian blue in the double null embryo. bo, basioccipitalelement; cv, first cervical vertebra; st, sternum; h, hyoid cartilage; c, cricoid cartilage; and t, thyroid cartilage.(C) Thoracic region. In the control embryo, ribs extended from the vertebrae to the sternum. They consisted of mature cartilage and growthplates in the ventral region and endochondral bone in the dorsal region (r1, first rib). In the double null embryo, ribs did not elongate towardthe ventral region and failed to stain with alcian blue.(D) Humerus (hu) of the control embryo and entire forelimb (fl) of the Sox5�/�; Sox6�/� embryo. The control humerus consisted of cartilagegrowth plates flanking a central core of endochondral bone. The double-mutant humerus was very short and surrounded by a thick intramembra-nous bone collar.(E) Distal phalange in a control embryo and corresponding digital ray in a double null embryo. The control phalange was organizing growthplates with reserve (res.), proliferative (prol.), and hypertrophic (hyp.) chondrocyte zones. Prechondrocytes composed the double null digi-tal ray.(F) Tibia proximal growth plate for the control embryo and proximal half for the double mutant. Proliferating chondroblasts (prol.), hypertrophicchondrocytes (hyp), and endochondral bone (bone) are shown in the control tibia. In the double null tibia, chondroblast-like cells were enlargingand accumulating a thin layer of cartilage matrix in the core region. Prechondrocytic cells were present in the epiphysis (epi) and surroundingthe diaphyseal core (double arrow).(G) Thoracic vertebral body. Control chondrocytes were fully hypertrophic. Double-mutant cells were also somewhat enlarged, but the cartilagematrix still poorly stained with alcian blue.(H) Control humerus proximal growth plate and entire double-mutant humerus. There were no cartilage growth plates and endochondral bone(single arrow) in the double mutant humerus, but a thick bone collar (double arrow) extended far along the epiphyses.(I) Basioccipital element. The picture shows the boundary between the endochondral bone (single arrow) and the hypertrophic chondrocytecartilage zone in the control embryo. There was no endochondral bone in the double mutant, but somewhat enlarged chondrocytes adjacentto a thick intramembranous bone surface (double arrow).(J) Sections directly adjacent to those shown in (I). Mineralization was seen in the ossified regions and around adjacent chondrocytes in bothcontrol and double null elements.

Page 10: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell286

Figure 6. Chondroblast-Deficient Functions in Sox5�/�; Sox6�/� Embryos

(A) Expression of cartilage matrix genes. Transverse sections through a thoracic vertebra of E15.5 Sox5�/�; Sox6�/� (left) and Sox5�/�; Sox6�/�

littermates (right) were hybridized with the indicated RNA probes. Pictures are centered on vertebral bodies, with neural tubes at the top. Indouble null cartilage, collagen 2, aggrecan, and link protein RNA levels were very low, and matrilin-1 and COMP RNA levels were undetectable.(B) Expression of perichondrium markers. The top panel shows transverse sections through the Meckel’s cartilage (arrow) of wild-type (left)and Sox5�/�; Sox6�/� (right) E14.5 littermates hybridized with a Gdf5 probe. M, molar tooth bud. The bottom panel shows longitudinal sectionsof the humerus of an E15.5 Sox5�/�; Sox6�/� embryo (left) and scapula of a Sox5�/�; Sox6�/� littermate (right) hybridized with a fibromodulinprobe. Strong hybridization signals were seen in wild-type perichondriums, and weak signals were seen in double null chondrocytes. Strongfibromodulin signals were seen in both wild-type and double null tendons (arrowheads).(C) Cell proliferation in wild-type E12.5 ribs. The left panels show alcian blue staining, and the right panels show PCNA staining. The toppanels are low-magnification pictures showing ribs growing from the dorsal side (d) toward the ventral side (v). The bottom panels are high-magnification pictures. In the dorsal area, chondroblasts were accumulating alcian blue-stainable matrix, and most were positive for PCNA(brown nuclei). In the ventral area, prechondrocytes were packed and all negative for PCNA. The dark blue staining of some small condensedcells corresponds to hematoxylin counterstaining but not PCNA staining.(D) Cell proliferation in a wild-type E16.5 humerus growth plate. At left, strong alcian blue staining in the periarticular reserve zone (res) andthe columnar and hypertrophic (col/hyp) zones is shown. The middle and right panels are high-magnification pictures of PCNA staining. Mostcells in the periarticular and columnar chondroblast regions stained intensely, whereas hypertrophic chondrocytes did not.

Page 11: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

L-Sox5 and Sox6 Control Chondrogenesis287

embryonic chondrogenesis. The sternum abnormality of Col11a2, since they bind to sequences needed for thecartilage-specific activity of enhancers present in theseSox6�/� mice was not severe enough to ascertain that

it was lethal. The postnatal dwarfism of Sox6�/� pups genes and cooperate with Sox9 to activate Col2a1 en-hancer constructs (Bridgewater et al., 1998; Lefebvre etsuggests a primary role for Sox6 in chondrogenesis

postnatally, but its origin could also be nonchondro- al., 1998; Zhou et al., 1998). Since other cartilage matrixgenes are coexpressed with Col2a1 and Col11a2 andgenic since Sox6 is expressed in several tissues (Le-

febvre et al., 1998). Hagiwara et al. (2000) reported that similarly downregulated in Sox5�/�; Sox6�/� embryos,it is likely that L-Sox5 and Sox6 also participate in theirmice homozygous for a chromosome 7 inversion that

disrupted Sox6 and the pink-eyed dilution p gene died direct activation. However, such evidence is presentlylacking, as the cis-acting elements controlling cartilageat birth or around postnatal day 14, like Sox6�/� mice.

Electrocardiograms and electron micrographs sug- expression of these genes are still unknown.The requirement of Sox5 and Sox6 for chondroblastgested skeletal and cardiac myopathy and heart block,

a phenotype not observed in p mutants. proliferation was demonstrated by the inability of dou-ble-mutant chondroblasts to resume proliferation forseveral days and then to maintain active proliferation inDistinct Functions for Sox5; Sox6 and Sox9epiphyseal cartilages. These data are consistent within Prechondrocytes and Chondroblaststhe close association between cell proliferation andSox5�/�; Sox6�/� embryos showed no defects in theSox5 and Sox6 expression. The two genes are indeedpatterning and early development of cartilages. Pre-

chondrocytes formed precartilaginous condensations in activated as growth-arrested prechondrocytes differen-the correct sites, in time, and in normal cell numbers. tiate into proliferating chondroblasts and inactivated asThey growth arrested, turned off mesenchymal markers, chondroblasts undergo postmitotic prehypertrophy. Fur-and turned on early cartilage markers. Since Sox9 is ther studies are, however, needed to determine whetherrequired for this early step (Bi et al., 1999), these data L-Sox5 and Sox6 directly control expression of cell cyclereveal that Sox9 is essential earlier than L-Sox5 and genes or whether their role is mediated through theirSox6 in chondrogenesis and is able to activate gene action on extracellular matrix production.expression independently of L-Sox5 and Sox6. Accord- Sox5�/�; Sox6�/� chondroblasts activated Cbfa1 andingly, Sox9 was activated in wild-type embryos at the other prehypertrophic and hypertrophic chondrocytetime of prechondrocyte emergence, Sox5 and Sox6 just markers in a manner that can be considered as preco-afterwards, and Sox9 was expressed at normal levels cious or ectopic. Similar phenotypic changes were in-in Sox5�/�; Sox6�/� prechondrocytes. These data thus duced upon forced expression of Cbfa1 in chondro-also identify Sox9 as a potential activator of Sox5 and blasts of transgenic mice (Takeda et al., 2001; Ueta etSox6, but rule out the possibility that L-Sox5 and Sox6 al., 2001). These data thus raise the possibility thatcontrol Sox9. L-Sox5 and Sox6 might act not only as transcriptional

Sox5�/�; Sox6�/� prechondrocytes were unable to enhancers but also as transcriptional repressors ofovertly differentiate into chondroblasts. They main- Cbfa1 and other late-stage chondrocyte markers. Theirtained low expression levels of early cartilage matrix role on these genes might, however, be indirect. Inter-genes, virtually failed to turn on stage-specific matrix estingly, Cbfa1 was correctly turned off in Sox5�/�;genes, resumed proliferation after a long delay, and then Sox6�/� prechondrocytes (data not shown), thus rulingrapidly activated late-stage chondrocyte markers. Al- out a role for L-Sox5 and Sox6 in Cbfa1 repression atthough Sox9 continues to be expressed in chondro- this early step.blasts, its role at this stage has not been demonstrated. The molecular mode of action of L-Sox5 and Sox6 isNevertheless, it is reasonable to assume that Sox9 con- still unknown, as is that of Sox13, their only close relativetinues to control Col2a1, aggrecan, and other genes (Bowles et al., 2000). Sox13 is not expressed in cartilagein chondroblasts, as it does in prechondrocytes. Even (Roose et al., 1998; our unpublished data) and thus can-though L-Sox5; Sox6 may cooperate with Sox9 in gene not account for the residual activation of cartilage matrixupregulation, they must act on these genes with different genes seen in Sox5�/�; Sox6�/� embryos. Therefore,molecular roles than Sox9, since they are structurally these data and the lack of transactivation domain invery different from Sox9 and significantly differ from L-Sox5 and Sox6 further suggest that L-Sox5 and Sox6Sox9 in their DNA binding and transcriptional activities act as transcriptional enhancers of cartilage matrixin vitro (Lefebvre et al., 1998). genes, in contrast to Sox9, which is a transcriptional

activator. L-Sox5 and Sox6 may act as architecturalproteins, like the HMG box LEF-1 protein (Giese et al.,Target Genes and Transcriptional Roles

of L-Sox5 and Sox6 1995). Upon DNA binding, the Sox and LEF-1/TCF HMGbox domains force DNA bending, a property that wasAll tested cartilage matrix genes were severely downreg-

ulated in Sox5�/�; Sox6�/� embryos. L-Sox5 and Sox6 shown to promote transcription of the TCR-� gene byallowing interaction between proteins bound to DNA onlikely participate in the direct activation of Col2a1 and

(E) Absence of cell proliferation in a Sox5�/�; Sox6�/� E16.5 digital ray. On the left is alcian blue staining; PCNA staining is shown at right.The majority of cells were still prechondrocytic and PCNA negative.(F) Cell proliferation in Sox5�/�; Sox6�/� E16.5 Meckel’s cartilage. Alcian blue staining is shown on the left; PCNA staining is shown on theright. Cells were starting to deposit cartilage matrix, and most were PCNA positive.(G) Abnormal cell proliferation in the epiphysis of a Sox5�/�; Sox6�/� E16.5 humerus. On the left is a low-magnification picture of alcian bluestaining. The cartilage epiphysis (epi), bone collar (bc), and shoulder presumptive joint area (pja) are indicated. On the right is a high-magnification picture of PCNA staining. Most cells stained weakly or did not stain for PCNA.

Page 12: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell288

Figure 7. Chondrocyte Maturation Defects in Sox5�/�; Sox6�/� Embryos

Alcian blue staining and RNA in situ hybridizations of E16.5 humerus longitudinal sections. The top panel for each marker shows a wild-typehumerus proximal or distal growth plate (but elbow joint for PTHrP). An asterisk marks the prehypertrophic chondrocyte zone shown as areference for the growth plate. Chondroblasts are located above the asterisk, and hypertrophic chondrocytes are located below the asterisk.The bottom panels show Sox5�/�; Sox6�/� humerus with the proximal epiphysis at the top. Sections slightly deviated from the longitudinalaxis. Most were through the core region and both epiphyses, but some (FGFR3 and Ptc1) were more lateral, showing only the core regionand one epiphysis. The bracket indicates the proximal half of the humerus (but the distal half for FGFR3).

either side of a LEF-1 site. Like other Sox proteins, accumulation of a thin layer of cartilage extracellularL-Sox5 and Sox6 may also pair off with other transcrip- matrix. Indeed, core cells in the wild-type humerus aretion factors (Kamachi et al., 2000). already expressing high levels of Ihh and starting to

express Col10a1 at E12.5, that is, soon after the humerushas become cartilaginous (St-Jacques et al., 1999).Chondrocyte Maturation in AbsenceSox5�/�; Sox6�/� cells underwent prehypertrophy withof Chondroblast Overt Differentiationa delay of two to three days compared to wild-type cells,In contrast to chondroblast markers, Sox5�/�; Sox6�/�

but like wild-type cells, the first mutant cells expressingcells activated prehypertrophic and hypertrophic chon-PPR and Ihh were seen as soon as they had accumu-drocyte markers at approximately wild-type levels, prov-lated some extracellular matrix.ing that L-Sox5 and Sox6 are not needed for expression

The maintenance of chondroblasts in the reserve andof these markers and that chondroblast overt differentia-columnar zones of cartilage growth plates is controlledtion is not mandatory for chondrocyte further matura-by the Ihh/PTHrP regulatory loop. Ihh signaling stimu-tion. The second conclusion is not unexpected if onelates chondroblast proliferation and PTHrP signaling de-recalls that chondroblasts in the core of wild-type carti-

lage primordiums undergo prehypertrophy soon after lays chondrocyte prehypertrophy (Lanske et al., 1996;

Page 13: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

L-Sox5 and Sox6 Control Chondrogenesis289

which an IRES-lacZ-pA/loxP-flanked PGKneobpA (lacZ/neo) cas-Vortkamp et al., 1996; St-Jacques et al., 1999; Karp etsette was inserted into the fifth coding exon for L-Sox5 (amino acidsal., 2000). The first prehypertrophic chondrocytes initiate153–212, corresponding to the N terminus of the coiled-coil domain)this loop by producing Ihh, which then signals to periarti-and the second coding exon for Sox6 (amino acids 14–80). This

cular chondroblasts to express PTHrP. Sox5�/�; Sox6�/�cassette and the MC1tkpA (tk) selection cassette were previously

chondroblasts failed to form reserve and columnar described (Bi et al., 1999). AB-1 129/SvEv ES clones harboring aSox5 or Sox6 heterozygous mutation were screened by Southernzones despite expression of Ihh by prehypertrophic cellsblot analysis using Sox5 and Sox6 probes located outside the ho-and expression of PTHrP by periarticular cells. Expres-mology regions used for gene recombination. Mouse chimeras weresion of Ptc1 at a high level throughout cartilages furthergenerated by blastocyst injection of mutant ES cell clones. Mutantindicated that Ihh signaling was functioning. Therefore,mice were analyzed in the C57BL/6 � 129/SvEv genetic background

these data suggest a link between PTHrP signaling and at the F1 to F5 generations. For each gene, homozygous null miceL-Sox5 and Sox6. L-Sox5 and Sox6 may be directly derived from two independent ES cell clones exhibited identical

phenotypes. Routine mouse genotyping was performed by PCR.required for optimal PTHrP signaling to occur or maybe needed to activate genes needed for signaling to

Embryo Analysisoccur. Despite absence of an obvious cartilage matrixTotal RNA was isolated from whole embryos and analyzed on North-deficiency in PTHrP null (Karaplis et al., 1994) and PPRern blots with probes for L-Sox5, Sox6, and 18S RNAs as describednull mice (Lanske et al.,1996), PTHrP signaling may inpreviously (Lefebvre et al., 1998). Staining of whole embryos with

turn act to maintain Sox5 and Sox6 expression or in- X-gal and alcian blue (Bi et al., 1999) and skeletal preparations (Ottocrease the transcriptional activities of L-Sox5 and Sox6, et al., 1997) were performed as described. Sections of formalin-as it does for Sox9. Sox9 is phosphorylated by protein fixed, paraffin-embedded embryos were stained with hematoxylin

and eosin, alcian blue and nuclear fast red, or von Kossa reactionkinase A (PKA) in response to PTHrP, and this modifica-and nuclear fast red according to standard protocols. Tartrate-resis-tion increases the DNA binding affinity of Sox9 (Huangtant acid phosphatase staining was performed using a leucocyteet al., 2000, 2001). L-Sox5 and Sox6 feature a PKA con-acid phosphatase kit (Sigma). Immunostaining for PCNA was per-

sensus site conserved between the two proteins but formed using a kit (Zymed). RNA in situ hybridization of embryounrelated to those of Sox9 (amino acids RKGS, 109–112, sections was performed as described (Albrecht et al., 1997) usingin L-Sox5; Lefebvre et al., 1998). This site might be a 35S-labeled antisense probes. Pictures of hybridization signals were

taken with a red filter and superimposed with blue fluorescencetarget of PTHrP signaling.images of cell nuclei stained with the Hoechst 33258 dye.Interestingly, mice lacking any of the major cartilage

matrix components displayed very similar defects inAcknowledgmentschondrocyte maturation and organization of cartilage

growth plates to those seen in Sox5�/�; Sox6�/� em-We thank H. Akiyama, P. Ducy, K. Nakashima, Y. Mishina, M. Sirito,

bryos. These included Col2a1 null mice (Li et al., 1995), W. Shawlot, and D. Whitworth for helpful technical advice; A. Brad-cartilage matrix deficiency (cmd) mice (Rittenhouse et ley, K.S.E. Cheah, C.X. Deng, R. Fassler, J. Hecht, M. Hook, H.

Kronenberg, F. Luyten, M.P. Scott, T. Shinomura, E. Vuorio, M. Wa-al., 1978; Wai et al., 1998), in which aggrecan was virtu-kamiya, Z. Werb, and Y. Yamada for cells, plasmids, and probes;ally absent (Watanabe et al., 1994), and link protein nulland G. Karsenty and P. Ducy for critically reading the manuscript.mice (Watanabe and Yamada, 1999). Therefore, the es-This work was funded by NIH grant AR42919 to B.d.C. and R.R.B,sential roles of L-Sox5 and Sox6 in the spatial and tem-an Arthritis Foundation Investigator Award to V.L., and NIH grant

poral control of chondrocyte differentiation are likely AR46249 to V.L. DNA sequencing was performed by M.D. Andersonmostly indirect, mediated through their ability to pro- Cancer Center sequencing core facility, supported by NCI grant

CA16672.mote cartilage matrix formation.

Received November 14, 2000; revised April 20, 2001.Conclusion

ReferencesL-Sox5 and Sox6 have essential, mostly redundant rolesalong the chondrocyte differentiation pathway. They act Albrecht, U., Eichele, G., Helms, J.A., and Lu, H.C. (1997). Visualiza-as transcriptional enhancers to stimulate chondroblasts tion of gene expression patterns by in situ hybridization. In Molecularin assuming their major differentiation functions, that is, and Cellular Methods in Developmental Toxicology (Boca Raton,

FL: CRC Press, Inc.), pp. 23–48.expression of cartilage extracellular matrix genes andcell proliferation. L-Sox5 and Sox6 are not needed for Bell, D.M., Leung, K.K.H., Wheatley, S.C., Ng, L.J., Zhou, S., Ling,

K.W., Sham, M.H., Koopman, P., Tam, P.P.L., and Cheah, K.S.E.further differentiation of chondroblasts into prehypertro-(1997). SOX9 directly regulates the type-II collagen gene. Nat. Genet.phic and hypertrophic chondrocytes, but by promoting16, 174–178.cartilage matrix formation they ensure proper spatialBi, W., Deng, J.M., Zhang, Z., Behringer, R.R., and de Crombrugghe,and temporal maturation of chondrocytes in cartilageB. (1999). Sox9 is required for cartilage formation. Nat. Genet. 22,growth plates. They thus play a central role in endochon-85–89.

dral skeleton development.Bowles, J., Schepers, G., and Koopman, P. (2000). Phylogeny ofthe SOX family of developmental transcription factors based on

Experimental Procedures sequence and structural indicators. Dev. Biol. 227, 239–255.

Bridgewater, L.C., Lefebvre, V., and de Crombrugghe, B. (1998).Please refer to the supplemental Experimental Procedures atChondrocyte-specific enhancer elements in the Col11a2 gene re-www.developmentalcell.com/cgi/content/full/1/2/277/DC1 for de-semble the Col2a1 tissue-specific enhancer. J. Biol. Chem. 273,tails.14998–15006.

Cancedda, R., Descalzi Cancedda, F., and Castagnola, P. (1995).Generation and Genotyping of Sox5 and Sox6 Mutant MiceChondrocyte differentiation. Int. Rev. Cytol. 159, 265–358.Sox5 and Sox6 genomic clones were isolated from a mouse 129/

SvEv genomic DNA library. Targeting vectors were constructed in DeLise, A.M., Fischer, L., and Tuan, R.S. (2000). Cellular interactions

Page 14: Developmental Cell, Vol. 1, 277–290, August, 2001 ... · lar, and most craniofacial bones are laid down through tion in some cartilages (Komori et al., 1997; Otto et al., a process

Developmental Cell290

and signaling in cartilage development. Osteoarthritis Cartilage 8, Koopman, P. (1997). SOX9 binds DNA, activates transcription, andcoexpresses with type II collagen during chondrogenesis in the309–334.mouse. Dev. Biol. 183, 108–121.Ducy, P., Zhang, R., Geoffroy, V., Ridall, A.L., and Karsenty, G. (1997).Otto, F., Thornell, A.P., Crompton, T., Denzel, A., Gilmour, K.C.,Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation.Rosewell, I.R., Stamp, G.W., Beddington, R.S., Mundlos, S., Olsen,Cell 89, 747–754.B.R., et al. (1997). Cbfa1, a candidate gene for cleidocranial dyspla-Giese, K., Kingsley, C., Kirshner, J.R., and Grosschedl, R. (1995).sia syndrome, is essential for osteoblast differentiation and boneAssembly and function of a TCR alpha enhancer complex is depen-development. Cell 89, 765–771.dent on LEF-1-induced DNA bending and multiple protein-proteinRittenhouse, E., Dunn, L.C., Cookingham, J., Calo, C., Spiegelman,interactions. Genes Dev. 9, 995–1008.M., Dooher, G.B., and Bennett, D. (1978). Cartilage matrix deficiencyHagiwara, N., Klewer, S.E., Samson, R.A., Erickson, D.T., Lyon, M.F.,(cmd): a new autosomal recessive lethal mutation in the mouse. J.and Brilliant, M.H. (2000). Sox6 is a candidate gene for p100H myop-Embryol. Exp. Morphol. 43, 71–84.athy, heart block, and sudden neonatal death. Proc. Natl. Acad. Sci.Roose, J., Korver, W., Oving, E., Wilson, A., Wagenaar, G., Markman,USA 97, 4180–4185.M., Lamers, W., and Clevers, H. (1998). High expression of the HMGHuang, W., Zhou, X., Lefebvre, V., and de Crombrugghe, B. (2000).box factor Sox-13 in arterial walls during embryonic development.Phosphorylation of SOX9 by cyclic AMP-dependent protein kinaseNucleic Acids Res. 26, 469–476.A enhances SOX9’s ability to transactivate a Col2a1 chondrocyte-St-Jacques, B., Hammerschmidt, M., and McMahon, A.P. (1999).specific enhancer. Mol. Cell. Biol. 20, 4149–4158.Indian Hedgehog signaling regulates proliferation and differentiationHuang, W., Chung, U.I., Kronenberg, H.M., and de Crombrugghe,of chondrocytes and is essential for bone formation. Genes Dev.B. (2001). The chondrogenic transcription factor Sox9 is a target of13, 2072–2086.signaling by the parathyroid hormone-related peptide in the growthTakeda, S., Bonnamy, J.P., Owen, M.J., Ducy, P., and Karsenty, G.plate of endochondral bones. Proc. Natl. Acad. Sci. USA 98, 160–(2001). Continuous expression of Cbfa1 in nonhypertrophic chon-165.drocytes uncovers its ability to induce hypertrophic chondrocyteJobert, A.S., Zhang, P., Couvineau, A., Bonaventure, J., Roume, J.,differentiation and partially rescues Cbfa1-deficient mice. GenesLe Merrer, M., and Silve, C. (1998). Absence of functional receptorsDev. 15, 467–481.for parathyroid hormone and parathyroid hormone-related peptideUeta, C., Iwamoto, M., Kanatani, N., Yoshida, C., Liu, Y., Enomoto-in Blomstrand chondrodysplasia. J. Clin. Invest. 102, 34–40.Iwamoto, M., Ohmori, T., Enomoto, H., Nakata, K., Takada, K., et al.Kamachi, Y., Uchikawa, M., and Kondoh, H. (2000). Pairing SOX off:(2001). Skeletal malformations caused by overexpression of Cbfa1with partners in the regulation of embryonic development. Trendsor its dominant negative form in chondrocytes. J. Cell Biol. 153,Genet. 16, 182–187.87–100.

Karaplis, A.C., Luz, A., Glowacki, J., Bronson, R.T., Tybulewikz,Vortkamp, A., Lee, K., Lanske, B., Segre, G.V., Kronenberg, H.M.,

V.L.J., Kronenberg, H.M., and Mulligan, R.C. (1994). Lethal skeletaland Tabin, C.J. (1996). Regulation of rate of cartilage differentiation

dysplasia from targeted disruption of the parathyroid hormone-by Indian hedgehog and PTH-related protein. Science 273, 613–622.

related peptide gene. Genes Dev. 8, 277–289.Wai, A.W.K., Ng, L.J., Watanabe, H., Yamada, Y., Tam, P.P.L., and

Karp, S.J., Schipani, E., St-Jacques, B., Hunzelman, J., Kronenberg,Cheah, K.S.E. (1998). Disrupted expression of matrix genes in the

H., and McMahon, A.P. (2000). Indian hedgehog coordinates endo-growth plate of the mouse cartilage matrix deficiency (cmd) mutant.

chondral bone growth and morphogenesis via parathyroid hormoneDev. Genet. 22, 349–358.

related-protein-dependent and -independent pathways. Develop-Watanabe, H., and Yamada, Y. (1999). Mice lacking link proteinment 127, 543–548.develop dwarfism and craniofacial abnormalities. Nat. Genet. 21,

Karsenty, G. (1998). Genetics of skeletogenesis. Dev. Genet. 22, 225–229.301–313.

Watanabe, H., Kimata, K., Line, S., Strong, D., Gao, L.Y., Kozak,Karsenty, G. (2001). Chondrogenesis just ain’t what it used to be. C.A., and Yamada, Y. (1994). Mouse cartilage matrix deficiencyJ. Clin. Invest. 107, 405–407. (cmd) caused by a 7 bp deletion in the aggrecan gene. Nat. Genet.Komori, T., Yagi, H., Nomura, S., Yamaguchi, A., Sasaki, K., Deguchi, 7, 154–157.K., Shimizu, Y., Bronson, R.T., Gao, Y.H., Inada, M., et al. (1997). Wegner, M. (1999). From head to toes: the multiple facets of SoxTargeted disruption of Cbfa1 results in a complete lack of bone proteins. Nucleic Acids Res. 27, 1409–1420.formation owing to maturational arrest of osteoblasts. Cell 89,

Xie, W.F., Zhang, X., Sakano, S., Lefebvre, V., and Sandell, L.J.755–764.(1999). Trans-activation of the mouse cartilage-derived retinoic acid-

Lanske, B., Karaplis, A.C., Lee, K., Luz, A., Vortkamp, A., Pirro, A., sensitive protein gene by Sox9. J. Bone Miner. Res. 14, 757–763.Karperien, M., Defize, L.H.K., Ho, C., Mulligan, R.C., et al. (1996).

Zhao, Q., Eberspaecher, H., Lefebvre, V., and de Crombrugghe, B.PTH/PTHrP receptor in early development and Indian hedgehog-(1997). Parallel expression of Sox9 and Col2a1 in cells undergoingregulated bone growth. Science 273, 663–666.chondrogenesis. Dev. Dyn. 209, 377–386.

Lefebvre, V., Li, P., and de Crombrugghe, B. (1998). A new long formZhou, G., Lefebvre, V., Zhang, Z., Eberspaecher, H., and de Crom-

of Sox5 (L-Sox5), Sox6 and Sox9 are coexpressed in chondrogen-brugghe, B. (1998). Three high mobility group-like sequences within

esis and cooperatively activate the type II collagen gene. EMBO J.a 48-base pair enhancer of the Col2a1 gene are required for carti-

17, 5718–5733.lage-specific expression in vivo. J. Biol. Chem. 273, 14989–14997.

Lefebvre, V., Huang, W., Harley, V.R., Goodfellow, P.N., and deCrombrugghe, B. (1997). SOX9 is a potent activator of the chondro-cyte-specific enhancer of the pro-alpha 1(II) collagen gene. Mol.Cell. Biol. 17, 2336–2346.

Leroy, J.G., Keersmaeckers, G., Coppens, M., Dumon, J.E., andRoels, H. (1996). Blomstrand lethal osteochondrodysplasia. Am. J.Med. Genet. 63, 84–89.

Li, S.-W., Prockop, D.J., Helminen, H., Fassler, R., Lapvetelainen,T., Kiraly, K., Peltarri, A., Arokoski, J., Lui, H., Arita, M., and Khillan,J.S. (1995). Transgenic mice with targeted inactivation of the Col2a1gene for collagen II develop a skeleton with membranous and peri-osteal bone but no endochondral bone. Genes Dev. 9, 2821–2830.

Ng, L.-J., Wheatley, S., Muscat, G.E.O., Conway-Campbell, J.,Bowles, J., Wright, E., Bell, D.M., Tam, P.P.L., Cheah, K.S.E., and