<<

Oral Science International 12 (2015) 1–4

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

Contents lists available at ScienceDirect provided by Elsevier - Publisher Connector

Oral Science International

j ournal homepage: www.elsevier.com/locate/osi

Review

The functions of Runx family transcription factors and Cbfb in

skeletal development

Toshihisa Komori

Department of Cell Biology, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan

a r a

t i c l e i n f o b s t r a c t

Article history: Runx family transcription factors consist of Runx1, Runx2, and Runx3. Runx1 is involved in chondro-

Received 9 October 2014

cyte differentiation at an early stage. Runx2 is a major for chondrocyte maturation,

Received in revised form 1 December 2014

and Runx3 has a redundant function with Runx2 and it is partly involved in chondrocyte maturation.

Accepted 4 December 2014

Runx2 directly regulates Ihh expression and enhances chondrocyte proliferation. Runx2 is an essential

transcription factor for osteoblast differentiation. Heterozygous mutation of RUNX2 causes cleidocranial

Keywords:

dysplasia, which is characterized by hypoplastic clavicles, open fontanelles, and supernumerary teeth.

Runx2

Runx2 directly regulates Sp7 expression, and Runx2 together with Sp7 and canonical Wnt signaling com-

Cbfb

Chondrocytes pletes determination of the lineage of mesenchymal cells into osteoblasts. Runx2 expression in osteoblasts

Osteoblasts is regulated by a 343-bp . Dlx5/6 and Mef2 directly bind this enhancer; form an enhanceosome

Enhancer with Tcf7, ␤-catenin, Sox5/6, Smad1, and Sp7; and activate this enhancer. Msx2 inhibits this enhancer,

and switching of the binding of Msx2 to Dlx5 is important for this activation. ␤ (Cbfb)

forms a heterodimer with Runx family and enhances their DNA-binding capacity. In Cbfb con-

ditional knockout mice in osteoblast and chondrocyte lineages, the differentiation of chondrocytes and

osteoblasts is inhibited. All of the Runx family proteins are reduced in the cartilaginous limb skeletons

and calvariae in Cbfb conditional knockout mice, although the reduction of Runx2 in calvariae is

much milder than that in cartilaginous limb skeletons. Therefore, Cbfb regulates skeletal development

by stabilizing Runx family proteins, and Runx2 protein stability is less dependent on Cbfb in calvariae

than in cartilaginous limb skeletons.

© 2014 Japanese Stomatological Society. Published by Elsevier Ltd. All rights reserved.

Contents

1. Introduction ...... 1

2. Runx2 and chondrocyte differentiation ...... 2

3. Runx2 and osteoblast differentiation ...... 2

4. Transcriptional regulation of Runx2 for osteoblast differentiation ...... 3

5. Cbfb and Runx family transcription factors ...... 3

6. Conclusions ...... 4

References ...... 4

1. Introduction then replaced with bone by osteoblasts and osteoclasts through

the process of endochondral ossification. After mesenchymal

The vertebrate skeleton is composed of cartilage and bone. cell condensation, pluripotent mesenchymal cells differentiate

Bone is formed through either intramembranous or endochondral into immature chondrocytes, which produce Col2a1 and proteo-

ossification. Osteoblasts directly form intramembranous bones, glycan. The immature chondrocytes mature into hypertrophic

while chondrocytes first form a cartilaginous skeleton, which is chondrocytes, which express Col10a1, and finally become ter-

minal hypertrophic chondrocytes, which express Spp1 (secreted

phosphoprotein 1/osteopontin), Ibsp (integrin-binding sialopro-

∗ tein/bone sialoprotein II), Mmp13 (matrix metallopeptidase 13),

Tel.: +81 95 819 7630; fax: +81 95 819 7633.

and Vegf (vascular endothelial growth factor) [1–3].

E-mail address: [email protected]

http://dx.doi.org/10.1016/S1348-8643(14)00032-9

1348-8643/© 2014 Japanese Stomatological Society. Published by Elsevier Ltd. All rights reserved.

2 T. Komori / Oral Science International 12 (2015) 1–4

Runx family transcription factors consist of Runx1, Runx2, and

Runx3. Runx2 is an essential transcription factor for osteoblast

differentiation [4,5]. Heterozygous mutation of RUNX2 causes clei-

docranial dysplasia (CCD), which is characterized by hypoplastic

+/−

clavicles, open fontanelles, and supernumerary teeth, and Runx2

mice also show hypoplastic clavicles and open fontanelles [6]. Fur-

thermore, Runx2 regulates chondrocyte maturation and Runx3 is

partly involved in it [2,7–12]. Runx2 is also required for vascular

invasion into the mineralized cartilage, which is composed of ter-

minal hypertrophic chondrocytes [13,14]. Runx1 is also involved in

chondrocyte differentiation, at least in the development of some of

the endochondral bones [15,16]. Core binding factor ␤ (Cbfb) is a

co-transcription factor, which forms a heterodimer with Runx fam-

ily transcription factors [17]. In this review, the major functions

of Runx2 in osteoblasts and chondrocytes, the functions of Cbfb

in skeletal development, and involvement of Runx1 and Runx3 in Fig. 2. Regulation of osteoblast differentiation by Runx2, Tcf/Lef, Ctnnb1, and Sp7.

Runx2, Tcf/Lef, Ctnnb1, and Sp7 are required for the differentiation of pluripotent

endochondral ossification are described.

mesenchymal cells to osteoblasts. Osteoblast-specific expression of Runx2 is reg-

ulated by Dlx5/6, Mef2, Tcf7, Ctnnb1, Smad1/5, Sp7, and Sox5/6, which activate

2. Runx2 and chondrocyte differentiation the 343-bp osteoblast-specific enhancer. Runx2 regulates Sp7 and Tcf7 expression.

Therefore, there are reciprocal regulations between Runx2 and Sp7 and between

Runx2 and Tcf7 [29].

Runx2 is weakly expressed in resting and proliferating chon-

drocytes and upregulated in prehypertrophic chondrocytes, and

maturation and vascular invasion into the cartilage (Fig. 1). How-

the high expression is maintained in hypertrophic and terminal

−/− ever, these findings also indicate that some transcription factors

hypertrophic chondrocytes [2]. Runx2 mice die at birth due to

have a redundant function with Runx2 in chondrocyte maturation.

the failure of breathing caused by the lack of ossification in the

−/− Runx3 is also expressed in prehypertrophic and hypertrophic

thoracic cage. In the skeleton of Runx2 newborn mice, all endo-

chondrocytes. Chondrocyte maturation is mildly retarded in

chondral bones are cartilaginous, and mineralization is virtually

−/−

Runx3 mice at E15.5 but not at E18.5. Hypertrophic chondro-

absent throughout the skeleton, except the tibia, fibula, radius,

cytes are completely absent, and the expression of Ihh and Pth1r

and ulna [4]. When the chondrocytes terminally differentiate, the

−/− −/−

is undetectable in the whole skeleton in Runx2 Runx3 mice,

matrix in the terminal hypertrophic chondrocyte layer is miner-

−/− indicating that chondrocyte maturation is completely blocked in

alized. In the tibia, fibula, radius, and ulna in Runx2 embryos

−/− −/−

Runx2 Runx3 mice [11]. Therefore, Runx2 is a major transcrip-

at embryonic day (E) 18.5, chondrocytes terminally differentiate

tion factor for chondrocyte maturation and Runx3 has a redundant

and express Spp1, Ibsp, and Mmp13, and the matrix is mineralized

function with Runx2 in chondrocyte maturation (Fig. 1).

without vascular invasion. However, chondrocyte hypertrophy and

−/−

The lengths of limbs are shortened in Runx2 mice and fur-

Col10a1 expression are virtually absent in the rest of the skeleton.

−/− −/−

ther shortened in Runx2 Runx3 mice [11]. As chondrocyte

Furthermore, the expression of Ihh and Pth1r is severely reduced

−/−

proliferation is reduced in Ihh mice, Ihh positively regu-

[2]. These findings indicate that Runx2 is required for chondrocyte

lates chondrocyte proliferation [18]. Ihh expression is severely

−/−

reduced in Runx2 mice. There are several perfect Runx2-binding

sequences in the Ihh , and Runx2 directly regulates Ihh

expression (Fig. 1). Although Ihh expression is undetectable in

−/− −/−

Runx2 Runx3 mice, Runx3 cannot regulate Ihh expression. The

−/− −/−

further reduction of limb length in Runx2 Runx3 mice com-

−/−

pared with that in Runx2 mice is due to the reduction in the size

of chondrocytes because chondrocyte maturation is completely

−/− −/−

blocked in Runx2 Runx3 mice [11].

3. Runx2 and osteoblast differentiation

Intramembranous bones and bone collar formation are com-

−/−

pletely absent in Runx2 mice. Osteoblast differentiation is

blocked at an early stage, and weak alkaline phosphatase activ-

ity is detected only in the periphery of the calcified cartilage in the

−/−

tibia, fibula, radius, and ulna [4]. Furthermore, Runx2 calvarial

cells spontaneously differentiate into adipocytes and differentiate

into chondrocytes in the presence of bone morphogenetic pro-

tein (BMP)-2 in vitro, but they do not differentiate into osteoblasts

even in the presence of BMP-2 in vitro or in vivo [19]. Therefore,

−/−

Runx2 mesenchymal cells have the potential to differentiate into

adipocytes and chondrocytes, but they completely lack the poten-

tial to differentiate into osteoblasts (Fig. 2).

−/−

Fig. 1. Regulation of chondrocyte maturation and proliferation by Runx2 and Osteoblast differentiation is also completely blocked in Sp7

Runx3. Runx2 enhances chondrocyte maturation and Runx3 is partly involved it. mice and mice with the deletion of Ctnnb1 (␤-catenin) in mes-

Runx2 induces the expression of Ihh, which enhances chondrocyte proliferation.

enchymal cells (Fig. 2). In both types of mice, perichondral

Ihh induces the expression of Pthlh, which inhibits Runx2 expression. Further, Ihh

mesenchymal cells, which express Runx2, condense and differen-

is required for the expression of Runx2 in mesenchymal cells in the perichondrium,

−/−

in which bone collar formation occurs. tiate into chondrocytes, indicating that Sp7 mesenchymal cells

T. Komori / Oral Science International 12 (2015) 1–4 3

osteoblasts. Furthermore, the histone variant H2A.Z is enriched

in the enhancer. These are typical chromatin modifications in

enhancers. The enhancer is sufficient to direct Runx2 expression

specifically to osteoblasts [29].

5. Cbfb and Runx family transcription factors

Inversion of 16 involves breakage of CBFB, and it

is associated with in humans. Cbfb forms

a heterodimer with Runx1, and both Runx1 and Cbfb are essential

−/−

for hematopoiesis in the fetal liver [30]. Cbfb mice die at midges-

tation due to the absence of fetal liver hematopoiesis. Rescue of

Fig. 3. The functions of Runx family transcription factors and Cbfb in skeletal devel-

−/−

opment. Runx1 and Runx2 have a redundant function in chondrocyte differentiation, hematopoiesis in Cbfb mice by mating with Cbfb transgenic

at least in some parts of the skeleton. Runx2 is a major transcription factor for mice under the control of Gata1 promoter, which directs reporter

chondrocyte maturation, and Runx3 is partly involved in it. Runx2 is an essential

to primitive and definitive erythroid cells, revealed

transcription factor for osteoblast differentiation, and the 343-bp enhancer regulates

the requirement for Cbfb in skeletal development [31]. Rescue of

Runx2 expression in osteoblasts. The contribution of Runx1 and Runx3 in osteoblast

−/−

hematopoiesis in Cbfb mice using Tek promoter Cbfb transgenic

differentiation remains to be clarified. Cbfb forms heterodimers with Runx family

transcription factors, enhances their DNA binding, and inhibits their degradation. mice also showed similar results [32]. In another mouse model in

which GFP was inserted into exon 5 of Cbfb to generate a Cbfb–GFP

fusion protein, the skeletal development was also impaired [33].

−/−

and Ctnnb1 mesenchymal cells maintain the potential to dif-

Chondrocyte maturation is delayed, chondrocyte proliferation

ferentiate into chondrocytes [20–23]. However, the mesenchymal

is reduced, and osteoblast differentiation is inhibited in Cbfb condi-

condensation and the differentiation of the mesenchymal cells into

tional knockout mice using Dermo1-Cre knock-in mice, in which Cre

−/−

chondrocytes are not observed in Runx2 mice [4]. As Sox9 is

recombinase is expressed in mesenchymal cells giving rise to both

essential for mesenchymal condensation and chondrocyte differ-

chondrocyte and osteoblast lineages [34]. The proteins of Runx1,

entiation during embryogenesis [24], these findings suggest that

Runx2, and Runx3 are reduced in the cartilaginous limb skeletons of

Runx2 is required for mesenchymal condensation, which is a pre-

Cbfb conditional knockout embryos at E15.5. The proteins of Runx1,

requisite for chondrocyte differentiation, in the cells that do not

Runx2, and Runx3 are also reduced in calvarial tissues of Cbfb con-

express enough Sox9.

ditional knockout embryos, although the reduction of the Runx2

Runx2 directly regulates Sp7 expression [25,26]. It also inhibits

protein in calvarial tissues is much milder than that in cartilagi-

adipocyte differentiation and directs the mesenchymal cells to

nous limb skeletons [34] (Fig. 3). Therefore, the Runx2 protein is

osteoblasts. Runx2 together with Sp7 and canonical Wnt signal-

more stable in calvariae than cartilaginous limb skeletons in the

ing completes the lineage determination of mesenchymal cells into

absence of Cbfb. This suggests that some unknown proteins com-

osteoblasts, completely blocking the ability of mesenchymal cells

pensate for the lack of Cbfb in the calvariae to protect against the

to differentiate into chondrocytes [27] (Fig. 2).

degradation of the Runx2 protein. Although the development of

+/−

endochondral bones is virtually normal in Runx2 mice, the cal-

+/−

4. Transcriptional regulation of Runx2 gene for osteoblast variae and clavicles are more severely affected in Runx2 mice

differentiation than in Cbfb conditional knockout mice [34]. The relative stability

of the Runx2 protein in the calvariae of Cbfb conditional knock-

Runx2 messenger RNA (mRNA) is transcribed from two pro- out mice can explain why Cbfb conditional knockout mice show a

moters, distal (P1) and proximal (P2) promoters, and specifically severe delay in endochondral ossification but milder deformities

expressed in osteoblasts and chondrocytes [28]. Runx2 expression in the calvariae and the lateral parts of clavicles, which are formed

in osteoblasts is mainly transcribed from the P1 promoter. How- through intramembranous ossification. All Runx family proteins are

ever, neither promoter drives reporter to osteoblasts in mice, involved in endochondral ossification, and Cbfb is likely to be nec-

suggesting the presence of an enhancer for the osteoblast-specific essary for all of the Runx family proteins in endochondral bone

expression. Green fluorescent protein (GFP) reporter mice using a formation (Fig. 3). In particular, Runx2 and Runx3 have redundant

bacterial artificial chromosome (BAC) clone of the Runx2 gene locus function in chondrocyte maturation, and Runx3 partly compen-

successfully recapitulated the endogenous expression of Runx2. sates for the lack of Runx2. Runx1 is involved in the development

An enhancer for osteoblast-specific expression has been identi- of sternum, occipital bone, and palate by regulating chondrocyte

fied by deletion of the BAC clone. GFP is specifically expressed in differentiation [15,16]. However, the impaired development of the

+/−

osteoblasts in GFP reporter mice driven by the 343-bp enhancer calvariae and clavicles in Runx2 mice indicates that Runx1 and

and a minimal promoter (Fig. 3). The sequence of this enhancer is Runx3 cannot compensate for the haplodeficiency of Runx2 in their

highly conserved among mouse, human, dog, horse, opossum, and development. It explains why CCD patients show typical pheno-

chicken. Dlx5/6 and Mef2 directly bind to the enhancer. Dlx5/6 and types in the calvariae and clavicles [34].

Mef2 form an enhanceosome with Tcf7, Ctnnb1, Sox5/6, Smad1, and Recently, other groups also reported various Cbfb conditional

Sp7, and they activate the enhancer (Fig. 2). Although both Msx2 knockout mice. Wei’s group generated Cbfb conditional knockout

and Dlx5 bind to the motif in the enhancer, Msx2 inhibits mice using Prrx1-Cre mice, Sp7-Cre mice, Col1a1-Cre mice, Dermo1-

the enhancer activity. The binding of Msx2 to the enhancer is Cre mice, and Col2a1-Cre mice [35–38]. They also revealed that Cbfb

detected in uncommitted mesenchymal cells, C3H10T1/2, whereas is required for chondrocyte maturation and osteoblast differentia-

the binding of Dlx5 to the enhancer is detected in osteoblastic tion and for both intramembranous and endochondral ossification.

MC3T3-E1 cells. Therefore, the switching of the binding from Msx2 However, they did not observe the reduction of Runx family pro-

fl/fl

to Dlx5 is important for activation of the enhancer [29]. teins in the cartilaginous skeletons of Cbfb Col2a1-Cre mice and

fl/fl fl/fl

The enhancer is highly enriched for histone H3 mono- and in the calvaria of Cbfb Sp7-Cre mice and Cbfb Col1a1-Cre mice

dimethylated at Lys4 and acetylated at Lys27 and Lys18, but [35,38]. Lim et al. also reported Cbfb conditional knockout mice

it is depleted for histone H3 trimethylated at Lys4 in primary using Col1a1-Cre mice [39]. They showed that cortical bone is

4 T. Komori / Oral Science International 12 (2015) 1–4

fl/fl

reduced in Cbfb Col1a1-Cre mice with the reduction in the Runx2 [14] Himeno M, Enomoto H, Liu W, et al. Impaired vascular invasion of Cbfa1-

deficient cartilage engrafted in the spleen. J Bone Miner Res 2002;17:1297–305.

protein, and that Cbfb inhibits polyubiquitination-mediated pro-

[15] Kimura A, Inose H, Yano F, et al. Runx1 and Runx2 cooperate during sternal

teosomal degradation of Runx2. The discrepancy in the stability of

morphogenesis. Development 2010;137:1159–67.

the Runx family proteins in these Cbfb conditional knockout mice [16] Liakhovitskaia A, Lana-Elola E, Stamateris E, et al. The essential requirement for

Runx1 in the development of the sternum. Dev Biol 2010;340:539–46.

may have been caused by the difference of the antibodies against

[17] Ogawa E, Inuzuka M, Maruyama M, et al. Molecular cloning and characteriza-

the Runx family proteins used for Western blot analyses.

tion of PEBP2 beta, the heterodimeric partner of a novel Drosophila runt-related

DNA binding protein PEBP2 alpha. Virology 1993;194:314–31.

[18] St-Jacques B, Hammerschmidt M, McMahon AP. Indian hedgehog signaling reg-

6. Conclusions

ulates proliferation and differentiation of chondrocytes and is essential for bone

formation. Genes Dev 1999;13:2072–86.

[19] Kobayashi H, Gao Y, Ueta C, et al. Multilineage differentiation of Cbfa1-deficient

Runx2 plays important roles in the differentiation of cells

calvarial cells in vitro. Biochem Biophys Res Commun 2000;273:630–6.

of two lineages, osteoblasts and chondrocytes. However, it was

[20] Nakashima K, Zhou X, Kunkel G, et al. The novel zinc finger-containing tran-

unknown how Runx2 transcription is regulated in osteoblasts scription factor osterix is required for osteoblast differentiation and bone

and chondrocytes. As the 343-bp enhancer is sufficient to induce formation. Cell 2002;108:17–29.

[21] Day TF, Guo X, Garrett-Beal L, et al. Wnt/beta-catenin signaling in mesenchy-

osteoblast-specific expression, this enhancer contains elements

mal progenitors controls osteoblast and chondrocyte differentiation during

essential to direct Runx2 expression to osteoblasts (Figs. 2 and 3).

vertebrate skeletogenesis. Dev Cell 2005;8:739–50.

Runx2 is the most upstream transcription factor for lineage com- [22] Hill TP, Spater D, Taketo MM, et al. Canonical Wnt/beta-catenin signal-

ing prevents osteoblasts from differentiating into chondrocytes. Dev Cell

mitment from mesenchymal stem cells to osteoblasts. Therefore, it

2005;8:727–38.

has also been revealed how mesenchymal stem cells are specified

[23] Hu H, Hilton MJ, Tu X, et al. Sequential roles of Hedgehog and Wnt signaling in

to the osteoblast lineage. Furthermore, it has been revealed that osteoblast development. Development 2005;132:49–60.

[24] Bi W, Deng JM, Zhang Z, et al. Sox9 is required for cartilage formation. Nat Genet

Cbfb plays important roles in skeletal development by stabilizing

1999;22:85–9.

Runx family transcription factors (Fig. 3).

[25] Nishio Y, Dong Y, Paris M, et al. Runx2-mediated regulation of the zinc finger

Osterix/Sp7 gene. Gene 2006;372:62–70.

References [26] Yoshida CA, Komori H, Maruyama Z, et al. SP7 inhibits osteoblast differentiation

at a late stage in mice. PLoS ONE 2012;7:e32364.

[27] Komori T. Signaling networks in RUNX2-dependent bone development. J Cell

[1] Marks SC, Odgren PR. Structure and development of the skeleton. In: Bilezikian

Biochem 2011;112:750–5.

JP, Raisz LG, Rodan GA, editors. Principles of bone biology. London: Academic

[28] Komori T, Kishimoto T. Cbfa1 in bone development. Curr Opin Genet Dev

Press; 2002. 1998;8:494–9.

[2] Inada M, Yasui T, Nomura S, et al. Maturational disturbance of chondrocytes in

[29] Kawane T, Komori H, Liu W, et al. Dlx5 and Mef2 regulate a novel Runx2

Cbfa1-deficient mice. Dev Dyn 1999;214:279–90.

enhancer for osteoblast-specific expression. J Bone Miner Res 2014;29:1960–9.

[3] Komori T. Regulation of bone development and extracellular matrix protein

[30] Komori T. Regulation of skeletal development by the Runx family of transcrip-

genes by RUNX2. Cell Tissue Res 2010;339:189–95.

tion factors. J Cell Biochem 2005;95:445–53.

[4] Komori T, Yagi H, Nomura S, et al. Targeted disruption of Cbfa1 results in a

[31] Yoshida CA, Furuichi T, Fujita T, et al. Core-binding factor beta interacts with

complete lack of bone formation owing to maturational arrest of osteoblasts.

Runx2 and is required for skeletal development. Nat Genet 2002;32:633–8.

Cell 1997;89:755–64.

[32] Miller J, Horner A, Stacy T, et al. The core-binding factor beta subunit is required

[5] Otto F, Thornell AP, Crompton T, et al. Cbfa1, a candidate gene for cleidocra-

for bone formation and hematopoietic maturation. Nat Genet 2002;32:645–9.

nial dysplasia syndrome, is essential for osteoblast differentiation and bone

[33] Kundu M, Javed A, Jeon JP, et al. Cbfbeta interacts with Runx2 and has a critical

development. Cell 1997;89:765–71.

role in bone development. Nat Genet 2002;32:639–44.

[6] Mundlos S, Otto F, Mundlos C, et al. Mutations involving the transcription factor

[34] Qin X, Jiang Q, Matsuo Y, et al. Cbfb regulates bone develop-

CBFA1 cause cleidocranial dysplasia. Cell 1997;89:773–9.

ment by stabilizing Runx family proteins. J Bone Miner Res 2014,

[7] Kim IS, Otto F, Zabel B, et al. Regulation of chondrocyte differentiation by Cbfa1.

http://dx.doi.org/10.1002/jbmr.2379 (in press).

Mech Dev 1999;80:159–70.

[35] Chen W, Ma J, Zhu G, et al. Cbfbeta deletion in mice recapitulates cleidocra-

[8] Enomoto H, Enomoto-Iwamoto M, Iwamoto M, et al. Cbfa1 is a positive

nial dysplasia and reveals multiple functions of Cbfbeta required for skeletal

regulatory factor in chondrocyte maturation. J Biol Chem 2000;275:8695–

development. Proc Natl Acad Sci U S A 2014;111:8482–7.

702.

[36] Fei T, Mengrui W, Lianfu D, et al. Core binding factor beta (Cbf() controls the

[9] Takeda S, Bonnamy JP, Owen MJ, et al. Continuous expression of Cbfa1 in

balance of chondrocyte proliferation and differentiation by upregulating Indian

nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic

hedgehog (Ihh) expression and inhibiting parathyroid hormone-related pro-

chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes

tein (PPR) expression in postnatal cartilage and bone formation. J Bone

Dev 2001;15:467–81.

Miner Res 2014;29:1564–74.

[10] Ueta C, Iwamoto M, Kanatani N, et al. Skeletal malformations caused by over-

[37] Wu M, Li C, Zhu G, et al. Deletion of core-binding factor beta (Cbfbeta) in mes-

expression of Cbfa1 or its dominant negative form in chondrocytes. J Cell Biol

enchymal progenitor cells provides new insights into Cbfbeta/Runxs complex

2001;153:87–100.

function in cartilage and bone development. Bone 2014;65:49–59.

[11] Yoshida CA, Yamamoto H, Fujita T, et al. Runx2 and Runx3 are essential for

[38] Wu M, Li YP, Zhu G, et al. Chondrocyte-specific knockout of cbfbeta reveals

chondrocyte maturation, and Runx2 regulates limb growth through induction

the indispensable function of cbfbeta in chondrocyte maturation, growth

of Indian hedgehog. Genes Dev 2004;18:952–63.

plate development and trabecular bone formation in mice. Int J Biol Sci

[12] Stricker S, Fundele R, Vortkamp A, et al. Role of Runx genes in chondrocyte

2014;10:861–72.

differentiation. Dev Biol 2002;245:95–108.

[39] Lim KE, Park NR, Che X, et al. Core binding factor beta of osteoblasts maintains

[13] Zelzer E, Glotzer DJ, Hartmann C, et al. Tissue specific regulation of

cortical bone mass via stabilization of Runx2 in mice. J Bone Miner Res 2014,

VEGF expression during bone development requires Cbfa1/Runx2. Mech Dev

http://dx.doi.org/10.1002/jbmr.2397 (in press). 2001;106:97–106.