当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 内分泌学杂志 > 2005年 > 第3期 > 正文
编号:11168209
Minireview: Transcriptional Regulation in Development of Bone
     Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114

    Address all correspondence and requests for reprints to: Henry Kronenberg, Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114.

    Abstract

    Regulation of gene expression by transcription factors is one of the major mechanisms for controlling cellular functions. Recent advances in genetic manipulation of model animals has allowed the study of the roles of various genes and their products in physiological settings and has demonstrated the importance of specific transcription factors in bone development. Three lineages of bone cells, chondrocytes, osteoblasts, and osteoclasts, develop and differentiate according to their distinct developmental programs. These cells go through multiple differentiation stages, which are often regulated by specific transcription factors. In this minireview, we will discuss selected transcription factors that have been demonstrated to critically affect bone cell development. Further study of these molecules will lead to deeper understanding in mechanisms that govern development of bone.

    BONE DEVELOPMENT IS achieved through the use of two distinct pathways, intramembranous and endochondral bone formation. In intramembranous bone formation, mesenchymal cells condense and directly differentiate into bone-forming osteoblasts. In contrast, in endochondral bone formation, mesenchymal cells condense and then become chondrocytes. This cartilage mold then directs the formation of osteoblasts, which form mature bone. This bone is continually remodeled by cycles of bone formation and resorption. Osteoclasts, of hematopoietic origin, are responsible for bone resorption. Thus, three distinct cell types (chondrocytes, osteoblasts, and osteoclasts) from two cell lineages direct the formation and remodeling of bone (1).

    Endochondral Bone Formation and Chondrocyte Differentiation

    Formation of the cartilage anlage starts with mesenchymal condensation (2). The molecular mechanism that regulates this process at the transcriptional level is not fully understood. However, because bone morphogenetic protein (BMP) signaling appears crucial for this process (3), Smads [a name derived from Sma and MADD (Mother against decapentaplegic)], transcription factors downstream of BMP signaling (4), likely play an important role. After the formation of mesenchymal condensations, cells start proliferating again and differentiate into chondrocytes (Fig. 1).

    FIG. 1. Differentiation of bone cells of three lineages and its regulation by transcription factors. Transcription factors important for bone cell differentiation are indicated by bold black letters with arrows at major differentiation steps. Signaling molecules are indicated by blue letters. Osteoblasts and chondrocytes are derived from common mesenchymal precursors. During endochondral bone formation, mesenchymal cells condense and differentiate into chondrocytes. BMP signaling is likely required for these processes. Sox9 and its related molecules L-Sox5 and Sox6 play a pivotal role in commitment and maintenance of chondrocyte phenotypes. In the growth plate, chondrocytes proliferate and further differentiate into column-forming cells, then into postmitotic hypertrophic chondrocytes. Runx2 expression stimulates terminal differentiation. Runx2 and Osx are both required for osteoblast differentiation. Runx2 expression starts before that of Osx. Its activity is regulated by interaction with other transcription factors such as members of the Twist family. Osteoclasts differentiate from hematopoietic lineage cells through multiple steps. M-CSF and RANLK are essential external stimuli for osteoclastogenesis. PU.1, MITFs, Nf-B, Fos/Fra1, and NFATc1 are all required for differentiation of mature osteoclasts.

    Sox9 (sex reversal Y-related high-mobility group box protein) and chondrocyte commitment

    Analysis of genetically manipulated mice demonstrated that Sox transcription factors, members of the high mobility group superfamily, are necessary for conversion of condensed mesenchymal cells to chondrocytes. Studies of chimeric mice (5) and conditional knockouts (6) have shown that Sox9 is required for formation of normal mesenchymal condensations, for conversion of mesenchymal cells to chondrocytes, for proliferation of chondrocytes, and for suppression of premature conversion of these chondrocytes to hypertrophic chondrocytes. Sox9 is also required for the production of L-Sox5 and Sox6, two related Sox family members required for normal chondrocyte function (7). Sox9 is not only required for development of chondrocytes but also directly regulates expression of genes important for chondrocyte function, such as Col2a1 (8), Col11a2 (9), Agc1 (aggrecan) (10), and Mia (Cdrap) (11). The function of Sox9 may be modulated by phosphorylation by the PTH-related peptide signaling pathway (12). It is, therefore, possible that Sox9 mediates part of PTH-related peptide action to regulate hypertrophic differentiation.

    Expression of Sox9 begins in the mesenchymal condensation. BMPs can induce SOX9 expression, and noggin, a BMP antagonist, blocks expression of SOX9 in mesenchymal condensations (13). Cytokines such as IL-1 and TNF-, and nuclear factor B (NF-B), the transcription factor that mediates many actions of these cytokines, suppress Sox9 expression, partly through induction of posttranscriptional mRNA degradation (14, 15); this suppression of SOX9 probably contributes to chondrocyte loss in inflammatory arthritis. How specific transcription factors regulate Sox9 gene transcription is a central question, although it appears complicated; genetic analysis of human and mouse Sox9 mutations demonstrated that cis elements quite distant from the transcription start site of the Sox9 gene are critical for Sox9 transcription in cartilage (16).

    Interestingly, overexpression of Sox9 in the cartilage causes a decrease in chondrocyte proliferation and a delay in bone development. This decrease in proliferation may result from binding of Sox9 to ?-catenin, the essential component of the canonical Wnt (a name that combines wingless and int) signaling pathway (17). Some Wnt members are expressed in cartilage, and effects of Wnt signaling in cartilage have been demonstrated (18, 19, 20, 21). Along with the observation that overactivation or deletion of ?-catenin in chondrocytes resulted in severe skeletal dysplasias, these findings suggest that the transcription complex, lymphoid enhancer-binding factor (LEF)/T-cell factor (TCF)/?-catenin, regulates some aspects of cartilage development (17).

    Runx2 and chondrocyte hypertrophy

    Chondrocytes divide and produce a characteristic matrix but then stop dividing, change the matrix they synthesize, and become quite large (hypertrophic). Runx2 and, to a lesser extent, Runx3, are the major transcription factors controlling the crucial steps because chondrocytes stop dividing and become hypertrophic. Runx transcription factors are members of the runt family, named for the DNA binding domain, conserved across species from Drosophila to humans (22). Mice missing Runx2 show a defect in chondrocyte maturation, with lack of hypertrophic chondrocytes in many bones (23), and mice missing both Runx2 and Runx3 completely lack chondrocytes (24). Furthermore, overexpression of Runx2 hastens hypertrophy and even converts chondrocytes in the trachea, which normally never hypertrophy, into hypertrophic chondrocytes and then bone (25, 26).

    The stages of chondrocyte differentiation are regulated by a complex series of signaling molecules and transcription factors in addition to Sox9 and Runx2/3. Differentiation of early periarticular chondrocytes into flat, columnar proliferating chondrocytes appears to be regulated by multiple signaling molecules such as Indian hedgehog (Ihh) and Wnts (19, 27). It is, therefore, likely that some of the downstream transcription factors triggered by Ihh and wnts, Gli family members, and the ?-catenin/TCF/LEF complex, play a role in this step. Chondrocyte differentiation is accompanied by changes in proliferation. Alterations in fibroblast growth factor (FGF) signaling (28, 29), BMP signaling (30), and Ihh signaling (31, 32, 33) participate in regulating chondrocyte proliferation. The suppressive effect of fibroblast growth factor signaling on proliferation is regulated by signal transducer and activator of transcription-1 at the transcriptional level (34). Overexpression of Ihh is associated with an increase in cyclin D1 expression (33), which ultimately stimulates cell cycle progression by activating E2F transcription factors (35). BMP signaling, therefore, involving Smad 1, 5, and 8 transcription factors, generally stimulates hypertrophic differentiation (30, 36, 37, 38, 39). Misexpression of a homeobox-containing transcription factor, Dlx5, which is normally expressed in the prehypertrophic region of the growth plate, stimulates hypertrophic differentiation in developing chicken limbs (40). Considering that Dlx5 is downstream of BMP signaling in several other cell types, Dlx5 and its related molecule, Dlx6 (41), may mediate BMP action regulating hypertrophic differentiation. These animal models with Dlx modulation, however, need cautious interpretation because neither misexpression nor deletion of the genes is chondrocyte specific.

    Osteoblast Differentiation

    Two transcription factors, Runx2 containing a runt domain (42, 43) and Osterix (Osx; SP7) with a zinc-finger motif (44) are absolutely required for osteoblast differentiation during both intramembranous and endochondral bone formation. Analysis of Osx null mice shows that Osx is genetically downstream of Runx2. Runx2 is expressed in the lateral mesoderm, mesenchymal condensations, and chondrocytes in addition to osteoblasts. Both overexpression of Runx2 and expression of a dominant-negative form of Runx2 in osteoblasts impairs bone formation, suggesting that regulation of different stages of osteoblast differentiation by Runx2 is complex (45, 46). Runx2 is regulated by phosphorylation and also interacts with other transcription factors, such as Smads 1 and 5 (47, 48), Smad 3 (49), signal transducer and activator of transcription-1 (50), Menin (51), Hey1 (52), Grg5 (53) p300 (54), and Twists (55). Runx2 target genes include genes expressed by mature osteoblasts, such as osteocalcin, bone sialoprotein, osteopontin, and collagen 1(I) (22).

    Little is known about how Osx regulates osteoblast differentiation and function. Expression of genes characteristic of mature osteoblasts is absent in cells surrounding chondrocytes in Osx null mice, and instead these cells express genes characteristic of chondrocytes. Thus, Osx may be important for directing precursor cells away from the chondrocyte lineage and toward the osteoblast lineage (44). Recently, an important role in osteoblast development for ?-catenin has become clear. ?-Catenin is the downstream mediator of canonical Wnt signaling that forms a transcription-regulating complex with TCF/LEF transcription factors. Inactivating mutations of LRP5 (low-density lipoprotein receptor-related protein 5), which encodes a Wnt coreceptor required for activation of canonical Wnt signaling, cause osteoporosis in humans (56) and in mice (57), whereas other mutations in LRP5 cause high bone mass (58, 59). Recent work in which ?-catenin is conditionally knocked out from cells at various stages of the osteoblast lineage, suggests that ?-catenin plays multiple critical roles in osteoblast differentiation (60, 61).

    Alterations in functions of various other non-bone-specific transcription factors have been also demonstrated to affect osteoblastic differentiation and function. These include activator protein-1 and its related molecules (62), Dlx5 (63, 64), Msx1 (65), Msx2 (66, 67, 68), Twist (69), Atf4 (70), and nuclear steroid hormone receptors such as androgen receptors (71) and estrogen receptors (72).

    Osteoclast Differentiation

    Osteoclasts develop from monocytic precursors of the hematopoietic lineage. Analysis of a variety of mutant mice with osteopetrosis, caused by loss or impairment of osteoclast function, has provided valuable insights into the genetic basis for osteoclast differentiation. The Ets family transcription factor, PU.1, is responsible for the earliest established event in osteoclastogenesis. PU.1 null mice lack not only osteoclasts but also macrophages, while preserving the ability to produce early monocytic cells (73). After commitment to the osteoclast lineage, mononuclear cells respond to macrophage colony-stimulating factor (M-CSF), produced by nearby stromal cells, through activation of c-fms, the receptor for M-CSF; mice with inactivating mutations of M-CSF form macrophages normally, but lack osteoclasts (74). The other signaling system essential for osteoclast differentiation is triggered when RANKL [receptor activator of nuclear factor B (RANK) ligand], a member of the TNF family, activates its receptor RANK, a member of the TNF receptor family. Several transcription factors have been found crucial for osteoclast differentiation downstream of M-CSF/c-fms and RANKL/RANK signaling. Microphthalmia-associated transcription factors (MITF), transcription factor E (TFE3), TFEB, TFEC, and MITF, are essential for differentiation of mononuclear precursors into multinucleated osteoclasts (75). The phenotypic variance among different mutations in the MITF gene and analysis of compound mutants for mitf and tfe3 demonstrated that these family members have redundant roles (76). MITF directly regulates genes important for osteoclast function such as tartrate-resistant acid phosphatase (TRAP) (77, 78), cathepsin K (79), and osteoclast-associated receptor (OSCAR) (80) and also may regulate other transcription factors essential for osteoclastogenesis, such as PU.1 and c-Fos, by physical interaction (77, 81). The importance of NF-B was demonstrated by that the absence of multinucleated bone-resorbing cells in NF-B mutant mice (82). Development of macrophages is preserved in NF-B null mice, suggesting that NF-B functions later than PU.1 during osteoclast differentiation. In addition to NF-B, the RANK signaling pathway activates at least two other transcription factors essential for osteoclast differentiation: c-fos, a member of the activator protein-1 family of transcription factors, and NFATc1 (nuclear factor of activated T cells, cytoplasmic, calcineurin-dependent 1). The absence of c-fos causes severe osteopetrosis with lack of osteoclasts (83, 84). This defect is rescued by expression of a Fos-related molecule Fra1that lacks a transactivation domain; this suggests that Fos regulates osteoclast formation by interacting with other DNA binding molecules (85). C-fos is required for the initial induction of NFATc1 expression (86, 87). NFATc1, identified as a gene up-regulated in RANKL-stimulated bone marrow-derived monocyte/macrophage precursor cells (88, 89), plays a critical role in osteoclast differentiation (90). Unlike wild-type ES cells, ES cells missing the NFATc1 gene are not able to differentiate into osteoclasts in vitro. Furthermore, overexpression of a constitutively active form of NFATc1 in c-fos null cells restores expression of osteoclast-specific genes, demonstrating that NFATc1 is a critical transcriptional regulator downstream of c-fos during osteoclast differentiation (87).

    We have briefly discussed transcription factors that have been shown through genetic studies to be important in bone development (see Fig. 1 for a summary of these transcription factors and critical signaling systems). Although animal models with genetic alterations demonstrate the physiological importance of genes, such models often give limited information about the primary targets of these transcription factors and subsequent cascades of gene expression. Therefore, the precise way that these transcription factors regulate bone cell development and function in vivo is limited and an important agenda for the future.

    References

    Karsenty G, Wagner EF 2002 Reaching a genetic and molecular understanding of skeletal development. Dev Cell 2:389–406

    Hall BK, Miyake T 2000 All for one and one for all: condensations and the initiation of skeletal development. Bioessays 22:138–147

    Pizette S, Niswander L 2000 BMPs are required at two steps of limb chondrogenesis: formation of prechondrogenic condensations and their differentiation into chondrocytes. Dev Biol 219:237–249

    Miyazawa K, Shinozaki M, Hara T, Furuya T, Miyazono K 2002 Two major Smad pathways in TGF-? superfamily signalling. Genes Cells 7:1191–1204

    Bi W, Deng JM, Zhang Z, Behringer RR, de Crombrugghe B 1999 Sox9 is required for cartilage formation. Nat Genet 22:85–89

    Akiyama H, Chaboissier MC, Martin JF, Schedl A, de Crombrugghe B 2002 The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev 16:2813–2828

    Smits P, Li P, Mandel J, Zhang Z, Deng JM, Behringer RR, de Crombrugghe B, Lefebvre V 2001 The transcription factors L-Sox5 and Sox6 are essential for cartilage formation. Dev Cell 1:277–290

    Lefebvre V, Huang W, Harley VR, Goodfellow PN, de Crombrugghe B 1997 SOX9 is a potent activator of the chondrocyte-specific enhancer of the pro 1(II) collagen gene. Mol Cell Biol 17:2336–2346

    Bridgewater LC, Lefebvre V, de Crombrugghe B 1998 Chondrocyte-specific enhancer elements in the Col11a2 gene resemble the Col2a1 tissue-specific enhancer. J Biol Chem 273:14998–15006

    Sekiya I, Tsuji K, Koopman P, Watanabe H, Yamada Y, Shinomiya K, Nifuji A, Noda M 2000 SOX9 enhances aggrecan gene promoter/enhancer activity and is up-regulated by retinoic acid in a cartilage-derived cell line, TC6. J Biol Chem 275:10738–10744

    Xie WF, Zhang X, Sakano S, Lefebvre V, Sandell LJ 1999 Trans-activation of the mouse cartilage-derived retinoic acid-sensitive protein gene by Sox9. J Bone Miner Res 14:757–763

    Huang W, Chung UI, Kronenberg HM, de Crombrugghe B 2001 The chondrogenic transcription factor Sox9 is a target of signaling by the parathyroid hormone-related peptide in the growth plate of endochondral bones. Proc Natl Acad Sci USA 98:160–165

    Healy C, Uwanogho D, Sharpe PT 1999 Regulation and role of Sox9 in cartilage formation. Dev Dyn 215:69–78

    Murakami S, Lefebvre V, de Crombrugghe B 2000 Potent inhibition of the master chondrogenic factor Sox9 gene by interleukin-1 and tumor necrosis factor-. J Biol Chem 275:3687–3692

    Sitcheran R, Cogswell PC, Baldwin Jr AS 2003 NF-B mediates inhibition of mesenchymal cell differentiation through a posttranscriptional gene silencing mechanism. Genes Dev 17:2368–2373

    Wunderle VM, Critcher R, Hastie N, Goodfellow PN, Schedl A 1998 Deletion of long-range regulatory elements upstream of SOX9 causes campomelic dysplasia. Proc Natl Acad Sci USA 95:10649–10654

    Akiyama H, Lyons JP, Mori-Akiyama Y, Yang X, Zhang R, Zhang Z, Deng JM, Taketo MM, Nakamura T, Behringer RR, McCrea PD, de Crombrugghe B 2004 Interactions between Sox9 and ?-catenin control chondrocyte differentiation. Genes Dev 18:1072–1087

    Guo X, Day TF, Jiang X, Garrett-Beal L, Topol L, Yang Y 2004 Wnt/?-catenin signaling is sufficient and necessary for synovial joint formation. Genes Dev 18:2404–2417

    Yang Y, Topol L, Lee H, Wu J 2003 Wnt5a and Wnt5b exhibit distinct activities in coordinating chondrocyte proliferation and differentiation. Development 130:1003–1015

    Hartmann C, Tabin CJ 2000 Dual roles of Wnt signaling during chondrogenesis in the chicken limb. Development 127:3141–3159

    Yamaguchi TP, Bradley A, McMahon AP, Jones S 1999 A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development 126:1211–1223

    Ducy P 2000 Cbfa1: a molecular switch in osteoblast biology. Dev Dyn 219:461–471

    Inada M, Yasui T, Nomura S, Miyake S, Deguchi K, Himeno M, Sato M, Yamagiwa H, Kimura T, Yasui N, Ochi T, Endo N, Kitamura Y, Kishimoto T, Komori T 1999 Maturational disturbance of chondrocytes in Cbfa1-deficient mice. Dev Dyn 214:279–290

    Yoshida CA, Yamamoto H, Fujita T, Furuichi T, Ito K, Inoue K, Yamana K, Zanma A, Takada K, Ito Y, Komori T 2004 Runx2 and Runx3 are essential for chondrocyte maturation, Runx2 regulates limb growth through induction of Indian hedgehog. Genes Dev 18:952–963

    Ueta C, Iwamoto M, Kanatani N, Yoshida C, Liu Y, Enomoto-Iwamoto M, Ohmori T, Enomoto H, Nakata K, Takada K, Kurisu K, Komori T 2001 Skeletal malformations caused by overexpression of Cbfa1 or its dominant negative form in chondrocytes. J Cell Biol 153:87–100

    Takeda S, Bonnamy JP, Owen MJ, Ducy P, Karsenty G 2001 Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes Dev 15:467–481

    Kobayashi T, Chung UI, Schipani E, Starbuck M, Karsenty G, Katagiri T, Goad DL, Lanske B, Kronenberg HM 2002 PTHrP and Indian hedgehog control differentiation of growth plate chondrocytes at multiple steps. Development 129:2977–2986

    Ornitz DM, Marie PJ 2002 FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev 16:1446–1465[Free Full Text]

    Iwata T, Li CL, Deng CX, Francomano CA 2001 Highly activated Fgfr3 with the K644M mutation causes prolonged survival in severe dwarf mice. Hum Mol Genet 10:1255–1264

    Minina E, Wenzel HM, Kreschel C, Karp S, Gaffield W, McMahon AP, Vortkamp A 2001 BMP and Ihh/PTHrP signaling interact to coordinate chondrocyte proliferation and differentiation. Development 128:4523–4534

    St-Jacques B, Hammerschmidt M, McMahon AP 1999 Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev 13:2072–2086

    Karp SJ, Schipani E, St-Jacques B, Hunzelman J, Kronenberg H, McMahon AP 2000 Indian hedgehog coordinates endochondral bone growth and morphogenesis via parathyroid hormone related-protein-dependent and -independent pathways. Development 127:543–548

    Long F, Zhang XM, Karp S, Yang Y, McMahon AP 2001 Genetic manipulation of hedgehog signaling in the endochondral skeleton reveals a direct role in the regulation of chondrocyte proliferation. Development 128:5099–5108

    Sahni M, Ambrosetti DC, Mansukhani A, Gertner R, Levy D, Basilico C 1999 FGF signaling inhibits chondrocyte proliferation and regulates bone development through the STAT-1 pathway. Genes Dev 13:1361–1366

    Coqueret O 2002 Linking cyclins to transcriptional control. Gene 299:35–55

    De Luca F, Barnes KM, Uyeda JA, De-Levi S, Abad V, Palese T, Mericq V, Baron J 2001 Regulation of growth plate chondrogenesis by bone morphogenetic protein-2. Endocrinology 142:430–436

    Tsumaki N, Nakase T, Miyaji T, Kakiuchi M, Kimura T, Ochi T, Yoshikawa H 2002 Bone morphogenetic protein signals are required for cartilage formation and differently regulate joint development during skeletogenesis. J Bone Miner Res 17:898–906

    Tsumaki N, Tanaka K, Arikawa-Hirasawa E, Nakase T, Kimura T, Thomas JT, Ochi T, Luyten FP, Yamada Y 1999 Role of CDMP-1 in skeletal morphogenesis: promotion of mesenchymal cell recruitment and chondrocyte differentiation. J Cell Biol 144:161–173

    Grimsrud CD, Romano PR, D’Souza M, Puzas JE, Reynolds PR, Rosier RN, O’Keefe RJ 1999 BMP-6 is an autocrine stimulator of chondrocyte differentiation. J Bone Miner Res 14:475–482

    Ferrari D, Kosher RA 2002 Dlx5 is a positive regulator of chondrocyte differentiation during endochondral ossification. Dev Biol 252:257–270

    Robledo RF, Rajan L, Li X, Lufkin T 2002 The Dlx5 and Dlx6 homeobox genes are essential for craniofacial, axial, and appendicular skeletal development. Genes Dev 16:1089–1101

    Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddington RS, Mundlos S, Olsen BR, Selby PB, Owen MJ 1997 Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell 89:765–771

    Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M, Sato M, Okamoto R, Kitamura Y, Yoshiki S, Kishimoto T 1997 Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 89:755–764

    Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, de Crombrugghe B 2002 The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108:17–29

    Liu W, Toyosawa S, Furuichi T, Kanatani N, Yoshida C, Liu Y, Himeno M, Narai S, Yamaguchi A, Komori T 2001 Overexpression of Cbfa1 in osteoblasts inhibits osteoblast maturation and causes osteopenia with multiple fractures. J Cell Biol 155:157–166

    Ducy P, Starbuck M, Priemel M, Shen J, Pinero G, Geoffroy V, Amling M, Karsenty G 1999 A Cbfa1-dependent genetic pathway controls bone formation beyond embryonic development. Genes Dev 13:1025–1036

    Lee KS, Hong SH, Bae SC 2002 Both the Smad and p38 MAPK pathways play a crucial role in Runx2 expression following induction by transforming growth factor-? and bone morphogenetic protein. Oncogene 21:7156–7163

    Zhang YW, Yasui N, Ito K, Huang G, Fujii M, Hanai J, Nogami H, Ochi T, Miyazono K, Ito Y 2000 A RUNX2/PEBP2 A/CBFA1 mutation displaying impaired transactivation and Smad interaction in cleidocranial dysplasia. Proc Natl Acad Sci USA 97:10549–10554

    Alliston T, Choy L, Ducy P, Karsenty G, Derynck R 2001 TGF-?-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. EMBO J 20:2254–2272

    Kim S, Koga T, Isobe M, Kern BE, Yokochi T, Chin YE, Karsenty G, Taniguchi T, Takayanagi H 2003 Stat1 functions as a cytoplasmic attenuator of Runx2 in the transcriptional program of osteoblast differentiation. Genes Dev 17:1979–1991

    Sowa H, Kaji H, Hendy GN, Canaff L, Komori T, Sugimoto T, Chihara K 2004 Menin is required for BMP-2-and TGF-?-regulated osteoblastic differentiation through interaction with Smads and Runx2. J Biol Chem 279:40267–40275

    Zamurovic N, Cappellen D, Rohner D, Susa M 2004 Coordinated activation of Notch, Wnt and TGF-? signaling pathways in BMP-2 induced osteogenesis: notch target gene Hey1 inhibits mineralization and Runx2 transcriptional activity. J Biol Chem 279:37704–37715

    Wang W, Wang YG, Reginato AM, Glotzer DJ, Fukai N, Plotkina S, Karsenty G, Olsen BR 2004 Groucho homologue Grg5 interacts with the transcription factor Runx2-Cbfa1 and modulates its activity during postnatal growth in mice. Dev Biol 270:364–381

    Sierra J, Villagra A, Paredes R, Cruzat F, Gutierrez S, Javed A, Arriagada G, Olate J, Imschenetzky M, Van Wijnen AJ, Lian JB, Stein GS, Stein JL, Montecino M 2003 Regulation of the bone-specific osteocalcin gene by p300 requires Runx2/Cbfa1 and the vitamin D3 receptor but not p300 intrinsic histone acetyltransferase activity. Mol Cell Biol 23:3339–3351

    Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz DM, Olson EN, Justice MJ, Karsenty G 2004 A twist code determines the onset of osteoblast differentiation. Dev Cell 6:423–435

    Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D, Zacharin M, Oexle K, Marcelino J, Suwairi W, Heeger S, Sabatakos G, Apte S, Adkins WN, Allgrove J, Arslan-Kirchner M, Batch JA, Beighton P, Black GC, Boles RG, Boon LM, et al 2001 LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 107:513–523

    Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass 2nd DA, Hartmann C, Li L, Hwang TH, Brayton CF, Lang RA, Karsenty G, Chan L 2002 Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol 157:303–314

    Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, Manning SP, Swain PM, Zhao SC, Eustace B, Lappe MM, Spitzer L, Zweier S, Braunschweiger K, Benchekroun Y, Hu X, Adair R, Chee L, FitzGerald MG, Tulig C, Caruso A, Tzellas N, Bawa A, Franklin B, McGuire S, et al. 2002 A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet 70:11–19

    Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Benichou O, Scopelliti D, Key L, Renton T, Bartels C, Gong Y, Warman ML, De Vernejoul MC, Bollerslev J, Van Hul W 2003 Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. Am J Hum Genet 72:763–771

    Hu H, Hilton MJ, Tu X, Yu K, Ornitz DM, Long F 2005 Sequential roles of Hedgehog and Wnt signaling in osteoblast development. Development 132:49–60

    Glass D, P.M, Long F, Taketo MM, McMahon AP, Karsenty G 2003 Regulation of bone formation by wnt signaling. J Bone Miner Res 18:S14 (Abstract)

    Sabatakos G, Sims NA, Chen J, Aoki K, Kelz MB, Amling M, Bouali Y, Mukhopadhyay K, Ford K, Nestler EJ, Baron R 2000 Overexpression of FosB transcription factor(s) increases bone formation and inhibits adipogenesis. Nat Med 6:985–990

    Acampora D, Merlo GR, Paleari L, Zerega B, Postiglione MP, Mantero S, Bober E, Barbieri O, Simeone A, Levi G 1999 Craniofacial, vestibular and bone defects in mice lacking the Distal-less-related gene Dlx5. Development 126:3795–3809

    Miyama K, Yamada G, Yamamoto TS, Takagi C, Miyado K, Sakai M, Ueno N, Shibuya H 1999 A BMP-inducible gene, dlx5, regulates osteoblast differentiation and mesoderm induction. Dev Biol 208:123–133

    Satokata I, Maas R 1994 Msx1 deficient mice exhibit cleft palate and abnormalities of craniofacial and tooth development. Nat Genet 6:348–356

    Liu YH, Tang Z, Kundu RK, Wu L, Luo W, Zhu D, Sangiorgi F, Snead ML, Maxson RE 1999 Msx2 gene dosage influences the number of proliferative osteogenic cells in growth centers of the developing murine skull: a possible mechanism for MSX2-mediated craniosynostosis in humans. Dev Biol 205:260–274

    Liu YH, Kundu R, Wu L, Luo W, Ignelzi Jr MA, Snead ML Maxson Jr RE 1995 Premature suture closure and ectopic cranial bone in mice expressing Msx2 transgenes in the developing skull. Proc Natl Acad Sci USA 92:6137–6141

    Satokata I, Ma L, Ohshima H, Bei M, Woo I, Nishizawa K, Maeda T, Takano Y, Uchiyama M, Heaney S, Peters H, Tang Z, Maxson R, Maas R 2000 Msx2 deficiency in mice causes pleiotropic defects in bone growth and ectodermal organ formation. Nat Genet 24:391–395

    Rose CS, Malcolm S 1997 A TWIST in development. Trends Genet 13:384–387

    Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC, Schinke T, Li L, Brancorsini S, Sassone-Corsi P, Townes TM, Hanauer A, Karsenty G 2004 ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry syndrome. Cell 117:387–398

    Yeh S, Tsai MY, Xu Q, Mu XM, Lardy H, Huang KE, Lin H, Yeh SD, Altuwaijri S, Zhou X, Xing L, Boyce BF, Hung MC, Zhang S, Gan L, Chang C, Hung MC 2002 Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci USA 99:13498–13503

    Sims NA, Clement-Lacroix P, Minet D, Fraslon-Vanhulle C, Gaillard-Kelly M, Resche-Rigon M, Baron R 2003 A functional androgen receptor is not sufficient to allow estradiol to protect bone after gonadectomy in estradiol receptor-deficient mice. J Clin Invest 111:1319–1327

    Tondravi MM, McKercher SR, Anderson K, Erdmann JM, Quiroz M, Maki R, Teitelbaum SL 1997 Osteopetrosis in mice lacking haematopoietic transcription factor PU.1. Nature 386:81–84

    Yoshida H, Hayashi S, Kunisada T, Ogawa M, Nishikawa S, Okamura H, Sudo T, Shultz LD 1990 The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature 345:442–444

    Hershey CL, Fisher DE 2004 Mitf and Tfe3: members of a b-HLH-ZIP transcription factor family essential for osteoclast development and function. Bone 34:689–696

    Steingrimsson E, Tessarollo L, Pathak B, Hou L, Arnheiter H, Copeland NG, Jenkins NA 2002 Mitf and Tfe3, two members of the Mitf-Tfe family of bHLH-Zip transcription factors, have important but functionally redundant roles in osteoclast development. Proc Natl Acad Sci USA 99:4477–4482

    Partington GA, Fuller K, Chambers TJ, Pondel M 2004 Mitf-PU.1 interactions with the tartrate-resistant acid phosphatase gene promoter during osteoclast differentiation. Bone 34:237–245

    Luchin A, Purdom G, Murphy K, Clark MY, Angel N, Cassady AI, Hume DA, Ostrowski MC 2000 The microphthalmia transcription factor regulates expression of the tartrate-resistant acid phosphatase gene during terminal differentiation of osteoclasts. J Bone Miner Res 15:451–460

    Motyckova G, Weilbaecher KN, Horstmann M, Rieman DJ, Fisher DZ, Fisher DE 2001 Linking osteopetrosis and pycnodysostosis: regulation of cathepsin K expression by the microphthalmia transcription factor family. Proc Natl Acad Sci USA 98:5798–5803

    So H, Rho J, Jeong D, Park R, Fisher DE, Ostrowski MC, Choi Y, Kim N 2003 Microphthalmia transcription factor and PU.1 synergistically induce the leukocyte receptor osteoclast-associated receptor gene expression. J Biol Chem 278:24209–24216

    Sato M, Morii E, Takebayashi-Suzuki K, Yasui N, Ochi T, Kitamura Y, Nomura S 1999 Microphthalmia-associated transcription factor interacts with PU.1 and c-Fos: determination of their subcellular localization. Biochem Biophys Res Commun 254:384–387

    Franzoso G, Carlson L, Xing L, Poljak L, Shores EW, Brown KD, Leonardi A, Tran T, Boyce BF, Siebenlist U 1997 Requirement for NF-B in osteoclast and B-cell development. Genes Dev 11:3482–3496

    Johnson RS, Spiegelman BM, Papaioannou V 1992 Pleiotropic effects of a null mutation in the c-fos proto-oncogene. Cell 71:577–586

    Wang ZQ, Ovitt C, Grigoriadis AE, Mohle-Steinlein U, Ruther U, Wagner EF 1992 Bone and haematopoietic defects in mice lacking c-fos. Nature 360:741–745

    Fleischmann A, Hafezi F, Elliott C, Reme CE, Ruther U, Wagner EF 2000 Fra-1 replaces c-Fos-dependent functions in mice. Genes Dev 14:2695–2700

    Rao A, Luo C, Hogan P.G 1997 Transcription factors of the NFAT family: regulation and function. Annu Rev Immunol 15:707–747

    Matsuo K, Galson DL, Zhao C, Peng L, Laplace C, Wang KZ, Bachler MA, Amano H, Aburatani H, Ishikawa H, Wagner EF 2004 Nuclear factor of activated T-cells (NFAT) rescues osteoclastogenesis in precursors lacking c-Fos. J Biol Chem 279:26475–26480

    Cappellen D, Luong-Nguyen NH, Bongiovanni S, Grenet O, Wanke C, Susa M 2002 Transcriptional program of mouse osteoclast differentiation governed by the macrophage colony-stimulating factor and the ligand for the receptor activator of NF B. J Biol Chem 277:21971–21982

    Ishida N, Hayashi K, Hoshijima M, Ogawa T, Koga S, Miyatake Y, Kumegawa M, Kimura T, Takeya T 2002 Large scale gene expression analysis of osteoclastogenesis in vitro and elucidation of NFAT2 as a key regulator. J Biol Chem 277:41147–41156

    Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J, Wagner EF, Mak TW, Kodama T, Taniguchi T 2002 Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell 3:889–901(Tatsuya Kobayashi and Hen)