Abstract
The major event that triggers osteogenesis is the transition of mesenchymal stem cells into bone forming, differentiating osteoblast cells. Osteoblast differentiation is the primary component of bone formation, exemplified by the synthesis, deposition and mineralization of extracellular matrix. Although not well understood, osteoblast differentiation from mesenchymal stem cells is a well-orchestrated process. Recent advances in molecular and genetic studies using gene targeting in mouse enable a better understanding of the multiple factors and signaling networks that control the differentiation process at a molecular level. Osteoblast commitment and differentiation are controlled by complex activities involving signal transduction and transcriptional regulation of gene expression. We review Wnt signaling pathway and Runx2 regulation network, which are critical for osteoblast differentiation. Many other factors and signaling pathways have been implicated in regulation of osteoblast differentiation in a network manner, such as the factors Osterix, ATF4, and SATB2 and the TGF-beta, Hedgehog, FGF, ephrin, and sympathetic signaling pathways. This review summarizes the recent advances in the studies of signaling transduction pathways and transcriptional regulation of osteoblast cell lineage commitment and differentiation. The knowledge of osteoblast commitment and differentiation should be applied towards the development of new diagnostic and therapeutic alternatives for human bone diseases.
Keywords: Osteoblast, Runx2, Osterix, ATF4, SATB2, Wnt signaling, TGF-Beta signaling, hedgehog signaling, fgf signaling, ephrin signaling, sympathetic signaling, Review
2. INTRODUCTION
Physiological bone turnover can be divided into 2 temporal phases: modeling, which occurs during development, and remodeling, a lifelong process involving tissue renewal. Remodeling starts with removal by osteoclasts of matrix, a mixture of insoluble proteins in which type I collagen is predominant (>90%) and a poorly crystalline, chemically modified hydroxyapatite. Following resorption, osteoblasts are recruited to the site, where they secrete and mineralize new matrix. The increased activity of osteoclasts caused by estrogen withdrawal causes bone loss and osteoporosis, a frequent low-bone mass disorder in postmenopausal women leading to structural instability and a high fracture risk. Estrogen deficiency is known to play a critical role in the development of osteoporosis, while calcium and vitamin D deficiencies and secondary hyperparathyroidism also contribute (1). Osteoporosis is a factor in more than 1.5 million fractures each year in the United States alone. Costs have been estimated at more than $17 billion a year, particularly from hip fractures, more than 75% of them in women. A better understanding of bone quality, coming from biochemical markers and refined imaging techniques, will help predict who is most at risk of debilitating fractures. One of the main approaches to gleaning details about the quality of bones is to measure the activity of osteoclasts and osteoblasts, the cells that remodel bone and thus influence its structural properties (2).
The osteopetrotic rodent models provided the first demonstration of contributions by osteoblasts to bone remodeling defects in osteopetrotic rats with specific genotypes (i.e., ia/ia, tl/tl, and op/op)(3–5). To understand the mechanisms of osteoblast differentiation, Ducy et al. initially studied the regulation of expression of Osteocalcin, the most osteoblast-specific gene (6). Over the past 12 years, molecular and genetic studies have modified our understanding of osteoblast differentiation (Table 1). Differentiation along the osteoblast lineage has been shown to depend on two transcriptional regulators. Runt-related transcription factor 2 (Runx2) and Osterix are both required for the early and late stages of osteoblast differentiation, whereby Runx2 is a master regulator that acts upstream of Osterix (7). Since the volume of Wnt literature is increasing rapidly, a few aspects of current interest have been selected here, mainly focused on Wnt signaling through its receptors (Frizzleds) to ß-catenin, which is often called the canonical pathway. The recent discoveries of signal transduction pathways and transcription factors critical for osteoblast differentiation and function have opened up new approaches to understanding the pathogenesis of osteoporosis. We focus this review on the process of osteoblast differentiation, mainly because this is the area in which much progress has recently been made.
Table 1.
Genes and their mouse models associated with impaired osteoblast function
Mutated Gene | Defects in Osteoblasts | Role | Reference Mouse Model |
---|---|---|---|
Runx2 | Devoid of osteoblasts and impaired final differentiation of chondrocytes | Functions as a master switch for inducing osteoblast differentiation | 173, 176 |
Runx2 interacting transcription factors | Role | Reference Mouse/Cell Model | |
AP1 | JunB is shown to be essential for osteoblast proliferation and differentiation; Most Fos proteins are also implicated in proliferation and differentiation of osteoblasts | 257 – 260 | |
Smad | Interacts with RUNX2 in vivo and in vitro and enhances the transactivation ability of this factor. The pathogenesis of CCD may be related to impaired Smad signaling | 90, 94, 225 | |
Ets1 | Runx2 and Ets1 cooperate in vivo to regulate expression of the Osteopontin (Opn) gene | 227 | |
C/EBPβ and −δ | C/EBP transcription factors support Osteocalcin gene expression and may play an important regulatory role during osteoblast differentiation | 228, 229 | |
Dlx5 | Has a critical positive role in osteoblast differentiation and subsequent mineralization. | 230,261 | |
Hes1 | Binds to and potentiates the transactivating function of Cbfa1 | 232 | |
Menin | Required for bone morphogenetic protein 2- and transforming growth factor beta-regulated osteoblastic differentiation through interaction with Smads and Runx2 | 233 | |
Dlx3 | Regulating osteoprogenitor cell differentiation and for both positive and negative regulation of gene transcription | 231 | |
Lef1 | Lymphocyte enhancer-binding factor 1 (Lef1) inhibits terminal differentiation of osteoblasts. | 262 | |
Msx2 | Stimulates the commitment of mesenchymal cells into an osteoblast lineage in association with inhibition of adipogenesis | 184 – 186 | |
PPARγ | Inhibits the Runx2-mediated transcription of osteocalcin in osteoblasts and terminal osteoblast differentiation | 238, 263 | |
Smad3 | Decreases Cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. | 239 | |
Hey1 | A negative regulator of osteoblast differentiation/maturation | 240 | |
Stat1 | Functions as a cytoplasmic attenuator of Runx2 in the transcriptional program of osteoblast differentiation | 241 | |
Osterix | Devoid of osteoblasts. | A critical transcription factor in osteoblast differentiation | 7 |
ATF4 | A delay in osteoblast differentiation. | Required for the timely onset and terminal differentiation of osteoblasts | 248 |
SATB2 | Defects in osteoblast differentiation. | Acts as a molecular node in a transcriptional network regulating osteoblast differentiation | 253 |
β-catenin | Blocked in differentiation and develops into chondrocytes instead. | Essential for osteoblast differentiation and preventing transdifferentiation of osteoblastic cells into chondrocytes | 67, 68, 264 |
3. OSTEOBLAST FUNCTION
Bone mass is regulated both by the number of mature osteoblasts and by their bone-forming activity. Osteoblasts are a bone-specific mesenchymal cell type, which is defined by its three functions. Firstly, it is responsible for bone formation, i.e., the synthesis and secretion of most proteins of the bone extracellular matrix (ECM), and also expresses genes that are necessary and sufficient to induce mineralization of this ECM. Osteoblasts deposit osteoid on the pre-existing mineralized matrix only. Osteoblasts ligate existing matrix via ß1 integrins, forming a monolayer that is linked by cadherins. Once active, the cells secrete a matrix containing type I collagen and smaller but significant amounts of osteocalcin, matrix gla protein (MGP), osteopontin, bone sialoprotein, many minor components, and, importantly, growth factors such as bone morphogenic proteins (BMPs) and transforming growth factor-ß (TGF-ß). Key ectoproteins, including progressive ankylosis gene (ANK) and tissue nonspecific alkaline phosphatase (TNAP), export pyrophosphate generated intracellularly and cleave this small-molecule inhibitor of calcification, respectively (8). Surprisingly, none of the genes involved in inducing ECM mineralization are osteoblast or even bone specific. However, osteoblast is the only cell type, along with odontoblasts and cementoblasts in teeth, in which they are coexpressed. A team led by Mary Schweitzer of North Carolina State University in Raleigh describes dinosaur blood vessels-still flexible and elastic after 68 million years-and apparently intact osteocytes (9). The unusual preservation of the originally organic matrix may be due in part to the dense mineralization of dinosaur bone, because a certain portion of the organic matrix within extant bone is intracrystalline and therefore extremely resistant to degradation. These factors, combined with as yet undetermined geochemical and environmental factors, presumably also contribute to the preservation of soft-tissue vessels.
In contrast to their proapoptotic action on osteoclasts, bisphosphonates increase the survival of osteoblastic cells (10). Despite its broad biological importance, the control of phosphate homeostasis remains incompletely understood. Most of the genes contributing to its normal regulation have been identified by studying genetic defects leading to different hypophosphatemic disorders. Dentin matrix protein 1 (DMP1) is a unique molecule that initiates osteoblast differentiation by transcription in the nucleus and orchestrates mineralized matrix formation extracellularly, at later stages of osteoblast maturation (11). Targeted ablation of both Dmp1 alleles in mice resulted in shorter bones and vertebrae, a highly expanded zone of proliferation and hypertrophic chondrocytes in the growth plate of younger mice and broad sclerotic long bones in older animals (12). These skeletal findings, which were initially thought to represent a form of chondrodysplasia, led to the conclusion that DMP1 is required for normal postnatal bone and tooth formation. Recently, however, decreased serum phosphate and calcium levels have been observed (13), demonstrating the similarity of this phenotype with different hypophosphatemia-induced forms of rickets. With the identification of DMP1 mutations as the cause of autosomal recessive hypophosphatemia (ARHP) (14), we have a better understanding of the regulation of phosphate homeostasis.
Secondly, osteoblasts are required for osteoclast differentiation, and thereby for bone resorption (15). The two main genes required for osteoclast differentiation, M-Csf and Rankl, are expressed in osteoblasts, and the requirement for osteoblasts to induce osteoclast differentiation can be bypassed by culturing osteoclast progenitor cells in the presence of M-CSF and RANKL. Recent studies by Elefteriou et al. provide interesting evidence that sympathetic signaling via ß2-adrenergic receptors increases expression of RANKL on osteoblast progenitor (16). Signaling by RANKL can be modulated by the decoy receptor Osteoprotegerin (Opg), which binds RANKL and is secreted from osteoblasts and several other cell types. Additionally, calcineurin/NFAT signaling in osteoblasts controls the expression of chemoattractants (CCL8, CCL6, and CCL12), which may recruit osteoclast precursors to bone and influence osteoclastogenesis (17).
Thirdly, bone and marrow are intrinsically linked with haematopoietic stem cells (HSCs), and their primitive progeny are located proximal to the endosteal surface of trabecular bone (18). Recently, bone morphogenetic protein receptor 1a (Bmpr1a) and parathyroid hormone (PTH)/PTH-related protein (PTHrP) receptor (PPR) studies identified osteoblasts as crucial cellular components of the HSC microenvironment and thus have defined a niche for HSCs (19, 20). These genetic studies provide mechanistic insights into osteoblast-mediated HSC expansion (21). The Bmpr1a studies identified a specific subset of N-cadherin-expressing osteoblasts that form an N-cadherin/ß-catenin adherens complex with HSCs, perhaps mediating the attachment or adhesion of HSCs within their niche (19). In the PPR studies, Notch signaling was implicated, because the Notch ligand Jagged 1 was highly expressed in osteoblasts and Notch-activated in HSCs (20). Subsequently, advanced imaging studies have demonstrated that the HSCs reside in close proximity to the bone-lining osteoblasts (22) as well as blood vessels, which may constitute an alternative niche (23).
The hematopoietic cytokine granulocyte-colony stimulating factor (G-CSF) is widely used clinically to elicit HSPC mobilization for life-saving BM transplantation. It has been postulated that G-CSF triggers the release of specific proteases, leading to the degradation of adhesion molecules and chemokines. In particular, CXCL12 or stromal-derived factor 1 (SDF-1) and its receptor CXCR4 have been implicated as a key ligand-receptor pair responsible for retention of HSCs in the bone marrow (24). However, the function of these proteases has been challenged by other data, indicating that G-CSF-induced mobilization was normal in mice lacking virtually all neutrophil serine protease activity, even when combined with a broad metalloproteinase inhibitor (25). This suggests that other proteases and/or other mechanisms are involved. Given that the ability to mobilize stem cells varies between patients, these processes beg further investigation. The work of Katayama et al. takes a step into this void, providing a mechanism for G-CSF-induced mobilization of HSCs and suggesting that the sympathetic nervous system may regulate the egress of stem and progenitor cells from their niche (26). Bone marrow, bone, and the nervous system now appear to integrate signals to regulate HSCs. If hematopoiesis is any guide, niches may be nodal points where multiple, previously disconnected systems collide (27).
4. OSTEOBLAST ORIGIN AND CELL LINEAGE
Osteoblasts, which play central roles in bone formation, are derived from undifferentiated mesenchymal cells that also have the capacity to differentiate into chondrocytes, adipocytes, and myoblasts (28). There are three major stages of osteoblastogenesis: proliferation, matrix maturation, and mineralization, which are characterized by sequentially expressed distinctive osteoblast markers. The most frequently used markers of the osteoblast differentiation process are alkaline phosphatase (ALP), type I collagen (Col1a1), osteopontin (OPN), bone sialoprotein (BSP), osteocalcin (OCN), and PPR (Table 2). In general, ALP, BSP and Col1a1 are early markers of osteoblast differentiation, while PPR and OCN appear late, concomitantly with mineralization. OPN peaks twice, during proliferation and then again in the later stages of differentiation.
Table 2.
Well-established markers of the osteoblast during developmental sequencing.
MSC | Immature Osteoprogenitor | Mature Osteoprogenitor | Preosteoblast | Differentiated osteoblast | Osteocyte |
---|---|---|---|---|---|
Alkaline Phosphatase (ALP) | − | + | ++ | +++ | − |
Phex | − | − | − | +++ | +++ |
Osteocalcin (OCN) | − | − | − | −→+++ | − |
Osteopontin (OPN) | −/+ | −/+ | −→+ | −→+++ | −→+++ |
Runx2 | + | + | ++ | +++ | +++ |
Osterix | − | − | ++ | ++ | ? |
Collα1 | − | ++ | ++ | ++ | − |
Bone sialoprotein (BSP) | −↔++ | ++↔− | −→+++ | −→+++ | −→+++ |
The list is not exhaustive, but does show some important categories of molecules in the lineage and their utility to help define transitions in osteoblast differentiation. −, no detectable expression; −/+, +, ++, +++, expression ranging from detectable to very high; −→+++, heterogeneous expression in individual cells.
Following initial lineage commitment, a phase of lineage expansion ensues which culminates normally in permanent cell cycle withdrawal. The initial cell division is asymmetric, giving rise to another stem cell (self-renewal) and a committed osteoprogenitor. Following commitment, the stem cell gives rise to the transit-amplifying compartment (29). This phase is associated with intensive proliferative activity. The preosteoblast is an intermediate stage, which expresses both STRO1, ALP, PPR, and type I collagen, and is committed to the osteoblast lineage with extensive replicative capacity, but no self-renewal capacity (30). In vitro, the use of agents such as retinoic acid can induce further differentiation in the preosteoblast. The mature osteoblast expresses ALP, OPN, BSP, and OCN, and lies adjacent to newly synthesized osteoid (Table 2). This stage, which is responsible for the laying down of bone, has limited replicative potential (31). The cumulative effect of the recruitment of stem cells and their expansion, and the functional capacity of mature osteoblasts, is measured by rates of bone formation in vivo. The second key step initiates terminal differentiation and permanent cell cycle withdrawal. The terminal stage of the bone lineage is the post-mitotic osteocyte, often found isolated within bone, presumably embedded within advancing osteoids. As an alternate fate, a proportion of cells in the transient amplifying compartment may also terminate in apoptosis.
A range of cytokines modulate osteoblast differentiation, including bone matrix-derived TGF-ß, bone morphogenic protein 2 (BMP-2), BMP-4, and BMP-7, and their inhibitors noggin, chordin, gremlin, and sclerostin, the last identified by positional cloning of families with increased bone mass. Similarly, numerous hormones impact osteoblast function positively including IGF-1, PTH, PTHrP, 1,25(OH)2D3, leptin, glucocorticoids, the Notch pathway, and members of the leukemia inhibitory factor/IL-6 family.
5. OSTEOBLAST SIGNALING
Several signaling systems are known to play important roles during osteoblast development (Figure 1).
Figure 1.
Six important signaling networks of osteoblast differentiation. Binding of Wnt to the FZD receptor induces ß-catenin accumulation, which translocates to the nucleus to activate target gene transcription. Several transcription factors have been found crucial for osteoblast differentiation downstream of this signaling pathway, such as Runx2, Osterix, and ATF4. They are essential for differentiation of mesenchymal stem cells into differentiated osteoblasts and also function in the transcription of osteoclast-specific genes. The dotted lines indicate that the physiological function or the stage at which the factor mainly works remains to be proven.
5.1 Wnt signaling pathway
5.1.1 Wnt signaling: an overview
Wnts, a family of secreted glycoproteins with multiple inhibitors, are ligands for the family of 7-membrane-spanning frizzled (FZD) receptors. The Wnt family of secreted factors is involved in numerous aspects of cellular biology, ranging from cell fate determination, polarity and differentiation to migration, proliferation, and function. Wnt proteins are divided into two classes. The first class activates the canonical Wnt signaling pathway, which involves the formation of a complex between Wnt proteins, FZD, and low density lipoprotein (LDL) receptor-related protein 5 (LRP5) or LRP6 receptors (32, 33). The noncanonical Wnt5a class binds FZD proteins, activates heterotrimeric G proteins, and increases intracellular calcium via protein kinase C-dependent mechanisms or induces Rho- or c-Jun N-terminal kinase (JNK)-dependent changes in the actin cytoskeleton (34).
Although Wnt proteins signal through several pathways to regulate cell growth, differentiation, function, and death, the Wnt/ß-catenin or canonical pathway appears to be particularly important for bone biology (35, 36). The complexities of the Wnt/ß-catenin signaling pathway in multiple cell types have been reviewed elsewhere (37, 38), and an outline of the current model of Wnt signal transduction is shown in Figure 1. Wnt proteins released from or presented on the surface of signaling cells act on target cells by binding to the FZD/LRP5/6 complex at the cell surface. Signals are generated through the proteins Disheveled, Axin, and Frat-1, which disrupt the protein complex and inhibit the activity of glycogen synthase kinase 3 (GSK3), thus causing hypophosphorylation of its substrate, ß-catenin (39) (Figure 1). So, the “on state” involves increasing the post-translational stability of ß-catenin, through WNT-dependent inhibition of GSK3 (Figure 1). Stabilized ß-catenin then accumulates in the cytosol and translocates to the nucleus to activate target gene transcription. The most studied nuclear partners of ß-catenin are the lymphoid enhancer-binding factor/T cell factor (Lef/Tcf) transcription factor family (40, 41). ß-catenin displaces corepressors of Lef/Tcfs (e.g., Groucho, silencing mediator of retinoid and thyroid receptors and nuclear receptor corepressor [SMRT/NCoR]), and forms heterodimers with the Lef/Tcf proteins. With the assistance of transcriptional coactivators (e.g., p300 and cAMP response element-binding protein [p300/CBP]), this heterodimer binds DNA and initiates the transcription of target genes (42).
Extracellular WNT ligands can interact with a host of secreted antagonists, including the secreted FZD-related protein (sFRP) family and Wnt inhibitory factor 1 (WIF-1; Figure 1), preventing activation of the pathway. LRP5/6 coreceptor activity is inhibited by members of the SOST (Sclerosteosis gene product)(43) and Dickkopf (Dkk) families (44), all of which bind LRP5/6. Both Dkk1 and Dkk2 antagonize canonical Wnt signaling by simultaneously binding to LRP5/6 (45) and a single-transmembrane protein called kremen (44). Li et al. found ample evidence that Dkk2 has a role in osteoblast terminal differentiation and the effects of Dkk2 may not be entirely mediated by its Wnt signaling antagonistic activity (46). The precise mechanisms by which Dkk2 is involved in terminal osteoblast differentiation need further investigation.
If Wnts are not expressed or if their binding to receptors is inhibited, degradation of ß-catenin is facilitated via interactions with a protein complex consisting of adenomatous polyposis coli (APC), axin, and GSK3. APC and axin act as scaffold proteins allowing GSK3 to bind and phosphorylate ß-catenin, identifying it for degradation by the ß-TrCP-mediated ubiquitin/proteasome pathway. In the nucleus, prospective target genes of the pathway are kept in a repressed state by interacting with T-cell factor (TCF) and LEF transcription factors, with associated corepressors. So, in the “off state,” cells maintain low cytoplasmic and nuclear levels of ß-catenin, although ß-catenin is associated with cadherins at the plasma membrane, an association that spares it from the degradative pathway (Figure 1) (41). Despite the implied importance of canonical Wnt signaling in osteoblast biology, the role of the TCF/LEF family of transcription factors is unclear. Since ß-catenin participates in transcriptional complexes with molecules other than TCF/LEFs, some target genes may not be regulated via TCF/LEF binding sites. For example, Kieslinger et al. found a strong synergy between LEF1 and EBF2, a member of the “early B cell factor”(EBF) family (47). Both EBF2 and LEF1/TCF proteins are expressed in specific cell types, and the functional synergy may contribute to a more restricted pattern of Wnt-regulated expression of Opg.
When considering WNT/ß-catenin signaling in disease, the potential involvement of multiple regulators of the effector ß-catenin must be included. Several components of the WNT/ß-catenin pathway might be regulated by WNT-independent processes. For example, there is evidence that integrin-linked kinase (ILK), a kinase that is regulated by integrin signaling, can inhibit GSK3, thereby stabilizing ß-catenin and activating ß-catenin target genes (48). Conversely, activation of the tumor suppressor p53 leads to degradation of ß-catenin (49). GSK3, as its name (glycogen synthase kinase) implies, is regulated by other pathways (50), raising questions about how signaling molecules are functionally compartmentalized. Finally, TGF-ß, Notch, and WNT pathways crosstalk in several contexts (34, 51, 52).
5.1.2 Wnt regulates osteoblastogenesis through the canonical pathway
Indisputably, Wnts are involved in embryonic skeletal patterning, fetal skeletal development, and adult skeletal remodeling (53–55). Unraveling the function(s) of Wnt proteins in the regulation of skeletogenesis has been a knotty problem, however, confounded by questions of functional redundancy, multiple times and sites of action, and the presence of other molecules that compete with Wnt function. The first indication that Wnt signaling plays a critical role in bone formation came from human studies where mutations in the Wnt coreceptor LRP5 are causally linked to alterations in human bone mass (56–59). These findings were supported by the observation that LRP5−/− mice also have low bone mass (60). Furthermore, gain-of-function mutations in LRP5 that increase Wnt signaling result in higher bone density in humans and mice (58, 61). Consistent with the effects of LRP5 on bone mass being mediated through canonical Wnt signaling, activation of this pathway in vitro results in the expression of alkaline phosphatase, an early osteoblast marker (62, 63).
One of the mechanisms whereby Wnt signaling increases bone formation is via stimulation of the development of osteoblasts, and there is considerable in vitro evidence supporting a role for Wnt/ß-catenin (i.e., canonical) signaling in this process (64–66). Higher levels of ß-catenin enhance bone formation with concomitant increases in expression of osteoblast-specific genes (64, 67), whereas conditional knockdown of the ß-catenin gene at an early developmental stage causes ectopic chondrogenesis and abnormal osteoblast differentiation (67–69). Clement-Lacroix et al. have shown that the GSK-3ß inhibitor LiCl increases bone formation in LRP5−/− mice (70).
Despite all of these data, the functions of Wnt signaling in the programs of bone biology remain unclear. Take, for example, two studies that show Wnt proteins inhibit the ability of human mesenchymal stem cells to differentiate into osteoblasts (71, 72) while three other studies show that Wnt signaling, through ß-catenin, contributes to osteoblast differentiation (35, 62, 73). The reason(s) for this apparent conflict are not immediately obvious. Most recently, Hill, T. P., et al; Day, T. F., et al; Glass, D. A., et al and Hu, H., et al provide compelling evidence that Wnt signaling represents both a cell-autonomous mechanism for inducing osteoblastic and suppressing chondrocytic differentiation in early osteochondroprogenitors and a mechanism in fully differentiated osteoblasts for stimulating the production of OPG, an inhibitor of osteoclast formation(67, 68, 74, 75).
5.1.3 Role of ß-catenin at various stages of osteoblast development
Among evolutionarily conserved signaling pathways, the pleiotropic effects of Wnt/ß-catenin signaling functions are well established in biological processes including embryogenesis, tumorigenesis, and stem cell biology (68, 76). Recent experiments examining the conditional inactivation of ß-catenin in skeletal progenitors and using different Cre lines revealed that ß-catenin activity is essential for the differentiation of mature osteoblasts and, consequently, for bone formation in endrochondral bones (the long bones of the limbs) and membranous bones (in the skull) (67, 68, 75). These variable results likely arise because Wnt/ß-catenin signaling regulates bone development and accrual through different mechanisms at different stages of life (77). This concept is supported by the results of studies using mouse models in which targeted deletion of ß-catenin occurs early or late in osteoblastogenesis.
Perichondrial and periosteal cells failed to express the osteoblast commitment factor, Osterix, and acquired a chondrogenic fate (67, 68). Similar to the long bones, the osteoblastic progenitors differentiated in the absence of ß-catenin into chondrocytes (67, 68). These findings were substantiated by in vitro deletion of ß-catenin activity in dissociated calvarial cells. It is likely that ß-catenin activity is required in a bipotential precursor of the osteoblast lineage, the so-called osteochondroprogenitor, and indeed its absence steers the fate of mesenchymal precursors toward chondrogenesis (67, 68). As Runx2, but not osterix, is expressed in ß-catenin−/− mesenchymal cells (68, 75), ß-catenin seems to be required for osteoblast differentiation at the preosteoblast stage (Table 1). Further, ß-catenin/TCF1 enhances Runx2 expression and Runx2 promoter activity (78). By contrast, for differentiation into the chondrocyte lineage, ß-catenin levels must be low (Figure 2) (67, 68, 75).
Figure 2.
Regulation of osteoblast differentiation by transcription factors. In osteoblast differentiation, high levels of Runx2 and ß-catenin are necessary to suppress the chondrogenic potential of uncommitted progenitors, such as the proposed osteochondroprogenitor. Osterix is required for the final commitment of progenitors to preosteoblasts. † signs indicate positive effects; ‡ signs indicate inhibitory effects.
Recently, a novel role for canonical Wnt signaling in postnatal bone homeostasis has been discovered by inactivating ß-catenin function in more mature osteoblasts using a Col1a1- and an OCN-Cre line (69, 74). Mice deficient in ß-catenin develop osteopenia. By contrast, activation of ß-catenin function in osteoblasts using the Col1a1- and the OCN-Cre line in combination with a conditional ß-catenin gain-of-function allele and a conditional APC allele, respectively, resulted in increased bone mass (69, 74). These mice manifest an osteopetrotic phenotype; however, no change in osteoblast activity or histomorphometric evidence of bone formation was observed. The altered bone resorption was caused by deregulation of Opg, a major inhibitor of osteoclast differentiation (74). Consistent with these observations in mice, autosomal-dominant osteopetrosis type I patients with a gain-of-function T253I mutation in LRP5 have decreased numbers of small osteoclasts, although osteoclastogenesis in response to RANKL was normal in vitro (79). Opg is a direct target gene of the ß-catenin-TCF complex in osteoblasts and Tcf1 is probably the relevant transcription factor required for Opg regulation; nevertheless, a possible role for Tcf4 cannot be excluded (65, 74). These mice demonstrate that ß-catenin regulates osteoclastogenesis through effects on expression of osteoprotegerin and RANKL (69).
5.2 TGF-beta Signaling
Once activated, TGF-ß can interact with its receptor to induce signaling. All members of the TGF-ß superfamily signal through a dual receptor system of type I and type II transmembrane serine/threonine kinases. The mothers against decapentaplegic (Smad) signaling turned out to play a central role in the transmission of signals from all receptors activated by the TGF-ß superfamily members to target genes in the nucleus.
Several members of the TGF-ß superfamily, such as BMPs, have potent osteogenic effects. BMPs are a group of phylogenetically conserved signaling molecules, and were initially identified by their capacity to induce endochondral bone formation (80–82). BMP-1 through BMP-7 are expressed in skeletal tissue, and BMP-2, -4 and -6 are the most readily detectable BMPs in osteoblast cultures (80, 83). BMPs are unique because they are implicated in the specification of both chondrocytes and osteoblasts (84), as well as in the subsequent modification of the osteogenic program, where some BMPs promote bone formation, such as BMP-2, BMP-7, BMP-6 and BMP-9 (85), although Bmp3 acts as a negative regulator of bone formation (86).
On receptor activation, BMPs transmit signals through Smad-dependent and Smad-independent pathways, including ERK, JNK, and p38 MAP kinase (MAPK) pathways (87). Smads are the major signal transducers for the serine/threonine kinase receptors (88). There are three classes of Smads: 1) receptor-regulated Smads (R-Smads) that can be BMP activated, such as Smad 1, 5 and 8 (referred to as BR-Smads in this article), or TGF-ß activated, such as Smad 2 and 3 (TR-Smads); 2) common partner BMP and TGF-ß mediator Smads (Co-Smads), such as Smad 4; and 3) inhibitory Smads (I-Smads), such as Smad 6 and 7. Upon ligand stimulation and activation by type II receptors, type I receptors phosphorylate R-Smads, which in turn form complexes with Co-Smads (89) (Figure 1). The R-Smad/Co-Smad complexes then translocate into the nucleus and regulate transcription of target genes by interacting with various transcription factors and transcriptional co-activators or co-repressors. The third class of Smads, I-Smads, negatively regulates signaling by the R-Smads and Co-Smads. Runx2 and BR-Smads physically interact with each other upon activation of BMP signaling, and co-operatively regulate the transcription of target genes, leading to osteoblast differentiation of mesenchymal progenitor cells (90–92). BMP induces Runx2 expression in mesenchymal progenitor cells through the action of BR-Smads (93), and BR-Smads in turn interact with Runx2 and further induce osteoblastic differentiation. BMP does not directly induce the expression of Runx2 in mesenchymal cells (94), but it facilitates expression of Dlx5 in osteoblasts (95, 96), and Dlx5 then induces expression of Runx2 in osteoprogenitor cells.
Osteoprogenitor cells, e.g. C2C12 cells, have been widely used for the identification of BMP target genes during osteoblastic differentiation. Hey1 (also termed HesR1 and Herp2) and Tcf7 are transcription factors specifically expressed in osteoblast cells by BMP-2 treatment, and are involved in Notch and Wnt signaling, respectively (97). Using constitutively active BMP type I receptors, Korchynskyi et al. identified several genes as targets of BMP receptors in C2C12 cells, including transcription factors Hey1, ITF-2, and ICSBP (98).
Although the Smads are critical mediators in the TGF-ß signaling pathway, a substantial body of evidence illustrates the existence of additional, Smad-independent pathways. BMP-2 can activate ERK, JNK and p38 in osteoblastic cells and provide evidences that these MAP kinases have distinct roles in regulating alkaline phosphatase and osteocalcin expression (99, 100). Recent reports suggest that during osteoblast differentiation, BMP-2 activates JNK and p38 via protein kinase D (PKD), independent of protein kinase C (PKC) activity (101). It has been demonstrated that following TGF-ß and BMP induction, both the Smad and p38 MAPK pathways converge at the Runx2 gene to control mesenchymal precursor cell differentiation (102). Runx2 plays a central role in the BMP-2-induced trans-differentiation of C2C12 cells at an early restriction point by diverting them from the myogenic pathway to the osteogenic pathway (94, 103). It has been found that the homeobox gene Dlx5 is an upstream target of BMP-2 signaling and that it plays a pivotal role in stimulating the downstream osteogenic master transcription factor Runx2 (96). In turn, Runx2 acts simultaneously or sequentially to induce the expression of bone-specific genes that represent BMP-2-induced osteogenic trans-differentiation. However, inhibition of BMP signaling was shown to disrupt the ability of RUNX2 to stimulate osteoblast differentiation and transactivate an osteocalcin gene promoter-luciferase reporter in C3H10T1/2 cells (104). In conclusion, we can state that the JNK, ERK, and p38 MAPK pathways contribute considerably to the whole of TGF-ß-induced responses, but further characterization is needed to assess their importance in relation to the Smad-dependent and other TGF-ß-induced signaling pathways.
BMP-2 has been reported to induce Osterix (Osx) expression in mouse progenitor cells and chondrocytes (7, 105). Moreover, BMP-2-induced Osx expression is mediated by Dlx5 but is independent of Runx2 (106). In the bone microenvironment BMPs act in conjunction with other growth factors. Celil et al. identified the involvement of BMP-2 and IGF-I in mediating Osx expression in human mesenchymal stem cells (hMSCs) (107). The BMP-2-induced effect on Osx expression was mediated via p38 but not via Erk. Under osteogenic culture conditions, both Erk and p38 were involved in mediating Osx expression (107).
In the past several years, ubiquitin-mediated proteasomal degradation has been implicated in the regulation of BMP-2 and TGF-ß signaling pathways in various cell types (108, 109). Recently, Dupont, S. et al and Yamashita, M.et al highlighted the importance of this mechanism in regulating the in vivo effects of TGF-ß (110, 111). Dupont, S. et al redefined the role of a previously identified Smad1 ubiquitin ligase, Smurf-1 The absence of Smurf-1 causes the accumulation of MEKK2, resulting in activation of JNK, an event that is both necessary and sufficient for BMP sensitization in osteoblasts (110). Yamashita, M.et al identified and characterized a novel Smad4 ubiquitin ligase, Ectodermin (Ecto) and provided convincing evidence that Ecto represents the elusive determinant of ectoderm formation, acting as a critical inhibitor of all Smad-dependent TGF-ß signaling during vertebrate development (111).
5.3 Hedgehog signaling
Osteoblast progenitors can first be identified within the inner perichondrium adjacent to, and coincident with, the first appearance of hypertrophic chondrocytes. This tight linkage reflects a crucial role for Indian hedgehog (Ihh) signaling (112). Ihh is produced by prehypertrophic chondrocytes and appears to act directly on perichondrially located osteoblast progenitors to specify the osteoblast precursors (113, 114). The failure of activation of Runx2, a crucial early determinant of the osteoblast lineage, indicates that hedgehog (Hh) signaling acts to initiate an osteogenic program (115). Furthermore, Hh activates osteoblast development in a variety of mesenchymal and skeletogenic cell types in vitro (114, 116, 117). Genetic manipulation of Smo, which encodes an obligatory component of the Hh signaling pathway, has revealed that cells devoid of Smo, hence Hh signaling, fail to undergo osteoblast differentiation(114). Although Ihh signaling plays the crucial role in regulating the temporal and spatial program of early osteoblast commitment, Ihh doesn't play an on-going role beyond this stage(118). When smoothened (Smo) activity is removed in Osx1+ osteoblast precursors, normal bone secreting OcHigh osteoblasts are generated, and the endochondral skeleton at birth is indistinguishable from wild type(118). Whether this is also true in the adult is currently under investigation.
The interaction between Hh and Wnt signaling is probably complex. It has been demonstrated that nuclear localization of ß-catenin as well as expression of target genes for the Wnt canonical pathway were abolished in the perichondrium in Ihh−/− embryos (75). This could, among other possibilities, be due to the downregulation of Wnt expression in the absence of Hh signaling. Indeed, expression of Wnt9a and Wnt7b was either reduced or abolished in the perichondrium in Ihh−/− embryos. In addition, both genes were induced by Hh signaling in C3H10T1/2 cells (Figure 1). Alternatively, the Hh and Wnt signaling pathways could intersect intracellularly via common regulators such as Suppressor of fused [Su(fu)](119) and GSK3 (120, 121). Other pathways in addition to canonical Wnt signaling also contribute to Hh-induced osteogenesis. Of note, some groups reported that Hh-induced osteogenesis in C3H10T1/2 cells required BMP signaling (122, 123).
5.4 FGF signaling
The fibroblast growth factors (FGFs) are a family of secreted polypeptides that act through four related tyrosine kinase receptors (Fgfr1–Fgfr4) to regulate a plethora of developmental processes, and they are critical for the control of endochondral and intramembranous ossification (124).
Human diseases that manifest the precocious osseous obliteration of sutures, known as craniosynostosis, often result from gain-of-function mutations in FGF receptors 1–3 (Figure 1) (125, 126). Fgfrs 1–3 are expressed in the developing and mature skeleton in patterns suggestive of both unique and redundant function (124). In the developing growth plate, both Fgfr1 and Fgfr2 are expressed in condensing mesenchyme that will give rise to cartilage. Fgfr2 remains expressed in reserve chondrocytes and appears to be down regulated in proliferating chondrocytes, whereas Fgfr1 is expressed in hypertrophic chondrocytes. Later in development, Fgfr1 and Fgfr2 are both expressed in the perichondrium and periosteum, tissues that give rise to osteoblasts and cortical bone. In contrast to Fgfr1 and Fgfr2, Fgfr3 is prominently expressed in proliferating chondrocytes where it regulates cell growth and differentiation (127) and in differentiated osteoblasts where it regulates bone density and cortical thickness (128, 129). Mutations in Fgfrs account for many of the craniosynostosis and chondrodysplasia syndromes in humans (124, 130, 131).
Embryos lacking Fgfr1 (Fgfr1−/−) die shortly after gastrulation (132), necessitating a conditional knockout approach to address function later in development. Hypomorphic alleles of Fgfr1 or conditional inactivation of Fgfr1 prior to limb bud initiation affects digital patterning and the formation of some skeletal elements (133–135). Fgfr1 signaling in the osteoprogenitor cell normally acts to stimulate differentiation whereas it functions to suppress differentiation in differentiated osteoblasts. Thus, Fgfr1 signaling has stage-specific effects on osteoblast maturation (136).
The FGF ligands that signal to Fgfr1 in osteoblasts are not known; however, three FGFs (FGFs 2, 9 and 18) are likely candidates for this role. FGF9 and FGF18 are expressed in the perichondrium/periosteum, and mice lacking these FGFs show delayed ossification during mid-gestation skeletogenesis (137–139). FGF2 is expressed in periosteal cells and osteoblasts (140, 141) and adult FGF2−/− mice showed a loss of trabecular bone volume; however, no skeletal dysmorphology was reported in neonatal FGF2−/− mice (142). These observations suggest that FGFs 2, 9 and 18 may act alone or redundantly to regulate osteoblast activity and physiology and that FGFs 9 and 18 may constitute the predominant signals during embryonic development, whereas FGF2 may be more important during postnatal stages. Consistent with a role for FGF2 in more differentiated osteoblasts, bone marrow stromal cultures from FGF2−/− mice showed a significantly decreased ability to mineralize in vitro (142). It has been demonstrated that Runx2 is phosphorylated and activated by FGF2 via the MAPK pathway and suggests that FGF2 plays an important role in regulation of Runx2 function and bone formation (143).
Fgfr2−/− mice die at embryonic day 10.5 (E10.5), prior to skeletal development (144). The contribution of Fgfr2 signaling to skeletal development has been clarified to some extent by using splice form-specific knockouts and conditional gene deletion approaches in mice. These studies demonstrated that Fgfr2 positively regulates bone growth and the anabolic function of osteoblasts. The resulting phenotype of mice lacking mesenchymal Fgfr2 included skeletal dwarfism, decreased bone density, incomplete formation of the dorsal vertebrae and tarsal joint fusion (145, 146). Alternative splicing of Fgfr2 is tissue specific, resulting in epithelial variants (b splice forms) and mesenchymal variants (c splice forms) (147–149). Ligand binding studies demonstrate that mesenchymally expressed ligands such as FGF7 and 10, activate Fgfr2b, whereas FGF2, 4, 6, 8 and 9 activate Fgfr2c (150, 151).
As adults, Fgfr3−/− mice were osteopenic, suggesting a role for Fgfr3 signaling in differentiated, Fgfr3-expressing osteoblasts (128). Mice lacking either FGF18 or Fgfr3 exhibited expanded zones of proliferating and hypertrophic chondrocytes and increased chondrocyte proliferation, differentiation, and Indian hedgehog signaling. These data suggest that FGF18 acts as a physiological ligand for Fgfr3 (138). In addition, FGF18−/− mice had decreased endochondral and intramembranous bone formation suggesting that FGF18 positively regulates osteogenesis and or osteoblast function independent of Fgfr3 (138).
Two lines of evidence indicate that En1 regulates signaling mediated by Fgfrs. First, the activation ERK, normally restricted to the mature endosteal osteoblasts of wild-type calvarial bone, is severely impeded in En1 mutants. Second, En1 ablation results in loss of the FGF target gene Spry2 in ectoperiosteal osteoblasts. Furthermore, En1 may regulate alternative FGF-signaling effectors known to affect osteoblast differentiation, such as p38 MAPK or PKC (152–154). A precise temporal and spatial delineation of these intracellular pathways will enable a better understanding of how osteoblastic differentiation is coordinated by En1 and FGFs.
To study the effects of growth factors on hMSC, Kratchmarova et al. tested the effects of epidermal growth factor (EGF), platelet-derived growth factor (PDGF), FGF, and nerve growth factor (NGF) on cellular responses and observed that EGF and PDGF elicited the strongest responses (155). They also found that the differentiation of human mesenchymal stem cells into boneforming cells is stimulated by EGF but not PDGF. Mass spectrometry-based proteomics analysis demonstrated that more than 90% of these signaling proteins were used by both ligands, whereas the phosphatidylinositol 3-kinase (PI3K) pathway was exclusively activated by PDGF, implicating it as a possible control point. Indeed, chemical inhibition of PI3K in PDGF-stimulated cells removed the differential effect of the two growth factors, bestowing full differentiation effects onto PDGF.
5.5 Ephrin signaling
Ephrins have the capacity for bidirectional signaling. That is, when a cell expressing an ephrin receptor contacts a cell expressing an ephrin ligand, signals are transduced into both the ephrin receptor-expressing cell (forward signaling) and the ephrin ligand-expressing cell (reverse signaling). There are two classes of ephrins, the B class (ephrin B1 to B3) are ligands for EphB tyrosine kinase receptors (B1 to B6), whereas class A ephrins (A1 to A5) are ligands for GPI-anchored EphA receptors (A1 to A10) (156). In bone biology, ephrinB and EphB receptors control patterning of the developing skeleton (157), and disruption of ephrin signaling is implicated in a syndrome called craniofrontonasal syndrome (CFNS [MIM 304110]) (158). Zhao et al. now suggested that ephrin signaling is critical to the two-way communication between osteoclasts and osteoblasts (159). This bidirectional signaling is mediated by the transmembrane ephrinB2 ligand in osteoclasts and EphB4, a tyrosine kinase receptor, in osteoblasts (Figure 1). Using osteoblast-osteoclast co-culture assays, as well as loss- and gain-of-function studies in vitro, the authors demonstrated that reverse signaling from EphB4 in osteoblasts to ephrinB2 in osteoclast progenitors leads to the inhibition of osteoclast differentiation. On the other hand, EphB4 expression is constitutive and the forward signaling through EphB4 induces osteogenic regulatory factors, such as Dlx5, Osx, and Runx2, in calvarial osteoblasts, suggesting that EphB4 is at the top of the regulatory cascade during osteoblast differentiation. Zhao et al. demonstrated that forward signaling between the extracellular domains of ephrinB2 and EphB4 in osteoblasts stimulates their differentiation, a process that may be dependent on RhoA inactivation in osteoblasts. Expressing active RhoA in osteoblasts may block the ability of ephrinB to promote osteoblast differentiation. By contrast, McBeath et al., using human mesenchymal stem cells, suggest that active RhoA enhances osteoblast differentiation (Figure 1) (160). Therefore, the involvement of RhoA in EphB4 forward signaling will need to be confirmed by future pharmacological or genetic studies. This study establishes the concept that ephrin-Eph signaling contributes to bone homeostasis.
5.6 Sympathetic signaling
Neural control of bone metabolism, both trophic and atrophic, has been suggested by numerous experimental and clinical observations. Osteoblasts have been reported to express receptors for several neuropeptides, suggesting that they could indeed integrate multiple neuronal signals (161). Immunolabeling studies have revealed a close association between glutamate-, catecholamine-, or peptide-containing nerve fibers and osteoblasts or osteoclasts in the endosteum (162). Blockade of glutamate receptors was reported to reduce the DNA binding activity and expression of Runx2 in cultured osteoblasts (163). The effect of the sympathetic nervous system (SNS) on bone formation has only recently been elucidated using genetic models (164). These studies revealed that leptin induced bone loss through SNS-derived signals originating in the ventromedial hypothalamic nuclei (162).
Fu et al indicated that an important, new regulator of bone remodeling is the circadian cycle (165). The model that emerges from the results of Fu et al. suggests that signaling by ß2-adrenergic receptors first activates the transcription factor CREB (Figure 1). CREB in turn stimulates expression of clock genes, which mediate the antiproliferative function by inhibiting G1 cyclin expression, and AP1 genes, which stimulate proliferation of osteoblasts. The Fu et al.'s work provided evidence that the inhibition of osteoblast proliferation by clock proteins is the dominant effect. Pharmacological or genetic ablation of adrenergic neurotransmission indicates that norepinephrine (NE) signaling controls G-CSF-induced osteoblast suppression (26). Based on studies describing leptin-mediated neuronal control of osteoblast function (16, 162) and the fact that leptin and G-CSF receptors display a high degree of homology (166), it has been proposed that G-CSF signals directly in the hypothalamus through the leptin receptor (26).
Sequence-specific DNA-binding proteins are frequently encoded by gene families. Such proteins display highly conserved DNA-binding properties, yet are assumed to retain promoter selectivity. Yamamoto et al. addressed the factor-specificity issue with his update on the varied roles of the glucocorticoid receptor (GR) (167). They presented evidence that two ligands, the small nuclear hormone and the larger DNA molecule, act to direct the activities of GR. Most fascinating were the data showing that different hormone ligands could affect promoter selectivity (167).
6. OSTEOBLAST GENE TRANSCIRPATION: TRANSCRIPTION FACTORS THAT REGULATE OSTEOBLAST DIFFERENTIATION
Transcription factors that regulate osteoblasts include a range of homeodomain proteins: the activator protein (AP) family members Jun, Fos, and Fra, Smads, CCAAT/enhancer binding protein ß (C/EBPß) and C/EBPd, lymphoid-enhancing factor (a Wnt effector), twist, activating transcription factor 4, Runx2, and osterix, the last 3 of which are considered master genes for osteoblast differentiation (Figure 2).
Commitment of mesenchymal stem cells (MSCs) to tissue-specific cell types is orchestrated by transcriptional regulators that serve as “master switches.” A central regulator of bone formation is the Runx2 transcription factor which fulfills its role as a master regulatory switch through unique properties for mediating the temporal activation and/or repression of cell growth and phenotypic genes as osteoblasts progress through stages of differentiation (168). Though Runx2 is essential for osteoblast differentiation, this differentiation program also requires other genes, such as osterix, which encodes a transcription factor genetically “downstream” of Runx2 (15). Thus, multiple genes regulate Runx2 activity and the effectiveness of Runx2 in stimulating osteoblast formation. It is perhaps not surprising then that Runx2 expression in osteoblast precursors predates by several days the first evidence for osteoblast activity.
6.1 Runx2
Runx2 is a member of the Runx (Runt-related factors) family of transcription factors (previously known as the acute myeloid leukemia [AML] factor, polyomavirus enhancer binding protein 2 [PEBP2], and core binding factor [CBF]). The family members, Runx1 (also called PEBP2aB, CBFA2 and AML1), Runx2 (also called PEBP2aA, CBFA1 and AML3), and Runx3 (also called PEBP2aC, CBFA3 and AML2), are encoded by distinct unlinked genes but share a common DNA recognition motif (TGTGGT) and heterodimerize with the ubiquitous subunit CBFß for stable DNA binding(169). Their highly conserved DNA binding domain is homologous to that from the Drosophila segmentation gene runt(170). In addition to the Runx DNA binding domain, Runx2 contains an active transactivation domain, rich in glutamine and alanine residues, and activates the Osteocalcin and Col1a1 genes(171, 172). Thus, Runx2 is an initial marker of the osteogenic cell lineage.
Runx2 is expressed in the thymus and testes (in T-lymphocytes tendon), and abundantly expressed in calcified cartilage and bone tissues, and is absent from the brain, heart, lung, gut, or liver (173–175). The function of Runx2 in bone formation has been demonstrated by analyzing its role in regulating the expression of the principal osteoblast-specific genes and by studying Runx2 null mice (171, 173, 176). Targeted disruption of Runx2 results in the complete lack of bone formation by osteoblasts, revealing that Runx2 is essential for both endochondral and membranous bone formation (Table 1) (173). The haploinsufficiency of the Runx2 gene, which leads to cleidocranial dysplasia, a genetic disease in humans that is characterized by hypoplastic clavicles, large open spaces between the frontal and parietal bones of the skull, and other skeletal dysplasias, is caused by heterozygous mutations in the Runx2 gene (177). Moreover, Runx2 is sufficient to induce osteoblast differentiation. This is true in cell culture, where forced expression of Runx2 in skin fibroblasts leads to osteoblast-specific gene expression (171), and in vivo, since ectopic expression of Runx2 leads to endochondral ossification in parts of the skeleton that would normally never ossify (178, 179). Importantly, Runx2 may function as an inhibitor of proliferation of progenitors, thus providing a mechanism for regulating the transition from growth to a postproliferative stage as a component of cellular commitment to the osteogenic lineage (180). Thus, Runx2 may be expressed in early osteoprogenitors to induce a program of gene expression required for lineage determination and differentiation of mesenchymal cells (Figure 2). Finally, Runx2 is also required for osteoblast function beyond differentiation (181, 182). These functions, along with its role during hypertrophic chondrocyte differentiation and vascular invasion, identify Runx2 as the most pleiotropic regulator of skeletogenesis (15). The literature now embraces the concept that Runx2 functions as a scaffold for the interaction with coregulatory proteins at subnuclear foci to provide an architectural basis for accommodating the requirements of biological control.
6.1.1 Runx2 upstream proteins
Msx2, which encodes a homeobox-containing transcription factor, is expressed in osteoblasts during development. The role of Msx2 during skull formation was first uncovered by human genetic studies. Indeed, one syndrome characterized by increased bone formation around the cranial suture, Boston-type craniosynostosis, is caused by an activating mutation in MSX2 (183). Msx2 inactivation in mice causes a marked delay of ossification in the bones of the skull and an overall decrease in bone volume. This phenotype is accompanied by a downregulation of Runx2 expression, indicating that Msx2 directly or indirectly regulates Runx2 expression (184). Recently, Cheng et al.(185) and Ichida et al.(186) reported that a homeobox gene, the Msx2 gene, stimulates the commitment of mesenchymal cells into an osteoblast lineage in association with inhibition of adipogenesis (Figure 2). Bapx1, another homeobox protein encoding gene, is required for axial skeleton formation. In Bapx1-deficient mice Runx2 expression is downregulated in the axial skeleton (187), suggesting that Bapx1 is another activator of Runx2 expression. Beside defects in branchial arch patterning, Hoxa2−/− mice show an upregulation of the cartilage- and bone- specifying genes Sox9 and Runx2 (188).
Twist-1 (previously called Twist) and Twist-2 (previously called Dermo-1) encode vertebrate basic helix-loop-helix transcription factors homologous to Drosophila Twist, a mediator of dorsal-ventral patterning and mesoderm formation. Knockout of Twist-1 in mice leads to lethality at E10.5 due to failure of neural tube closure (189). Twist-1 heterozygotes (both in mice and in humans) exhibit craniosynostosis, a disease caused by premature osteoblast differentiation in the skull. Bialek et al. show that Runx2-induced osteoblast gene expression only occurs when expression of Twist genes disappears in osteoblast precursors (190). Twist-1 heterozygosity reverses skull abnormalities in Runx2+/− mice and Twist-2−/− reverses clavicular abnormalities in Runx2+/− mice and accelerates osteoblast differentiation in bones formed through endochondral bone formation. Twist proteins' antiosteogenic function is mediated by a novel domain, the Twist box, which interacts with the Runx2 DNA binding domain to inhibit its function. They conclude that Twist-1 and Twist-2 regulate the developmental action of Runx2 in bone formation through the direct interaction of these proteins. Bialek et al., by bringing together the actions of Twist and Runx proteins, have clarified an important stage in bone formation and set a research agenda for the future (191).
It has long been recognized that the p53 tumor suppressor plays a pivotal role in preventing cancer. Two independent studies (192, 193) have addressed the role of p53 in bone differentiation in mouse models. In one case, Wang et al. examined skeletal structure and bone metabolism in p53 knockout mice (193). Conversely, Lengner et al. analyzed the effects of hyperactive p53 on bone formation caused by the conditional deletion of Mdm2 in osteoblasts (192). Surprisingly, and in contrast to the in vitro studies (194), both groups came to the same conclusion that p53 suppresses differentiation. Specifically, p53−/− osteoblasts displayed a marked propensity to differentiate, which was manifested by a modest but significant increase in bone formation and bone density in adult p53 knockout mice. Consistent with these results, the conditional deletion of Mdm2 in osteoblasts interfered with terminal differentiation, leading to late stage embryonic lethality, where the embryos displayed more porous and shorter bones. These findings suggest that the interplay between p53 and Mdm2 could either positively or negatively impact bone development. The studies of both Lengner et al. and Wang et al. provide compelling evidence that p53 suppresses osteoblast differentiation by repressing the expression of either Runx2 or Osterix (Figure 2) (192, 193). The subtle discrepancy that exists between the two studies (whether Runx2 or Osterix is the target of p53 action) may be related to how p53 activity is targeted and whether this mechanism alters the stage of cell differentiation. In either case, the concept that the absence of a tumor suppressor gene can enhance cell proliferation while favoring the differentiation of mesenchymal stem cells is intriguing but counterintuitive. It is likely that p53-deficient osteoprogenitors can still exit the cell cycle upon terminal differentiation, which may be enhanced as a result of the elevated expression of Runx2 and Osterix. These findings clearly establish p53 as a negative regulator of osteoblast differentiation both in vitro and in vivo.
Schnurri-3 (Shn3), a large zinc finger protein, was originally identified as a DNA binding protein of the heptameric recombination signal sequence required for V(D)J recombination of immunoglobulin genes (195); however, it also functions as an adapter protein in the immune system (196). Jones et al. found that mice lacking Shn3 display adult-onset osteosclerosis with increased bone mass due to augmented osteoblast activity (197). Shn3 was found to control protein levels of Runx2 by promoting its degradation through recruitment of the E3 ubiquitin ligase WWP1 to Runx2. By this means, Runx2-mediated extracellular matrix mineralization was antagonized, revealing an essential role for Shn3 as a central regulator of postnatal bone mass (197). This area of osteoblast biology is still in its infancy, and the transcription factors that act upstream of Runx2 to control its expression remain to be identified.
6.1.2 Runx2 interacting proteins
6.1.2.1 Co-activators of Runx2
Runx2 protein is shown to interact with a number of transcriptional co-activators (Figure 2). The most important coregulatory protein, essential for enhancement of Runx DNA binding is Cbfß (also known as PEBP2ß), the non-DNA-binding partner of all three Runx proteins. Inactivation of Cbfß causes embryo lethality in mice between E11.5 and E13.5. This results from hemorrhaging in several tissues and the absence of liver hematopoiesis because Cbfß is a heterodimerizing partner of Runx1 and Runx3, which are essential for haematopoiesis. The timing of embryonic lethality precludes examining the role of the Cbfß subunit in osteoblast differentiation. Transgenic rescue of embryonic lethal Cbfß-null mice and `knock-in' of Cbfß fused in-frame to a cDNA encoding green fluorescent protein (198) resulted in mice that exhibited delayed ossification, indicating a role for Cbfß in bone. However, unlike Runx2-null mice that completely lack bone and osteoblasts, ossification is initiated in these mice, suggesting that Runx2 can act in the absence of Cbfß. Similar observations were made simultaneously by two other groups (199, 200). Runx2 also interacts physically and/or functionally with other well-characterized coactivators, including p300, CBP, MOZ, and MORF. p300 and CBP physically interact with various R-Smads upon ligand stimulation and enhance Smad-dependent transcription of target genes (201, 202). Neither p300 nor CBP has been coprecipitated with Runx2. Two members of the MYST family of HATs, MOZ and MORF, however, do interact with the activation domain of Runx2 and enhance activation of the osteocalcin promoter (203). This interaction may be functionally relevant to intramembranous bone formation, as mice with a mutation in the MYST gene have craniofacial defects (204). Grg5, pRb, and TAZ are other proteins that enhance Runx2-mediated transactivation (205–208). Although most Grg/TLE proteins are corepressors, Grg5 appears to be a dominantnegative form of longer Grg/TLE proteins, and thereby enhances Runx2 activity in vivo (207). The 14-3-3-binding protein, TAZ (transcriptional coactivator with PDZ-binding motif), coactivates Runx2-dependent gene transcription while repressing PPARg-dependent gene transcription, indicating that TAZ functions as a molecular rheostat to fine-tune the balance between osteoblast and adipocyte development (208).
6.1.2.2 Co-repressors of Runx2
Histone deacetylases (HDACs) remove acetyl groups from lysine residues on many proteins, including histones. The elimination of the acetyl group alters chromatin structure by removing a mark needed to recruit co-activating proteins and by facilitating chromatin condensation to promote transcriptional repression (209). General HDAC inhibitors, such as trichostatin A, increase Runx2-mediated activation, and Runx2 associates with several HDACs, including HDAC3, HDAC4, and HDAC6 (Figure 2) (210–212). HDAC3 interacts with the N terminus of Runx2. Suppression of HDAC3 expression in differentiating MC3T3-E1 cells accelerates matrix mineralization and the expression of bone marker genes such as osteopontin, bone sialoprotein, and osteocalcin (211). Vega et al. claimed that HDAC4 inhibits Runx2 activity by blocking Runx2 DNA binding (212). HDAC4-null mice display premature ossification due to early onset chondrocyte hypertrophy, and overexpression of HDAC4 inhibits chondrocyte hypertrophy, suggesting that Runx2 activity is controlled by HDAC4 in prehypertrophic chondrocytes (212). HDAC6 was identified as a Runx2 binding protein in co-immunoprecipitation experiments designed to identify co-repressors that bind to the potent C terminus repression domain of Runx2 (210). Moreover, several other groups have reported that HDAC inhibitors increased Runx2-dependent activation of the osteocalcin promoter (211) and osteoblast maturation and differentiation (213, 214). However, results from a new study support a requirement for HDAC4 and -5 deacetylase activity to regulate acetylation, abundance and activity of Runx2 (215). As the phenotypes of other HDAC mutations are probed in detail, it will be exciting to learn whether these proteins have a general role controlling osteoblast differentiation.
Grg/TLE proteins are coexpressed with Runx2 in skeletal cells (216). The induction of the OCN gene correlates with downregulation of the level of Grg/TLE in mice skeletal tissues between E14 and birth and Grg/TLEs were shown to inhibit Runx2 dependent activation of OCN gene transcription (217). However, Grg5, a dominant negative form of long Grg/TLE proteins, can enhance Runx2 transcriptional activity in vitro (207). Depletion of Grg5 alone, with normal activity of Runx2, causes postnatal growth retardation in about 50% of the mice, and in Grg5 null Runx2+/− mice, the lack of Grg5 function combined with the heterozygous loss of Runx2 activity resulted in a growth deficiency which was more pronounced than would have been expected. This finding suggests that Grg5 and Runx2 interact with each other in vivo and that their combined activity is necessary for the activation of another factor important for bone and cartilage development. It is highly probable that the factor regulated by Grg5-Runx2 interaction is Ihh (207). Runx2 also interacts with YAP, a mediator of Src/Yes signaling, in the cytoplasm and translocates it to the nuclear matrix where YAP represses Runx2-mediated activation of the osteocalcin promoter (218). It is not yet known whether Runx2 associates with multiple corepressor complexes, or whether all of the corepressors mentioned above are components of the same complex.
One of the mechanisms by which transcription factors are regulated is by modulation of degradation. The proteosome degradation pathway decreases Runx2 protein levels to slow osteoblast differentiation (219). The ubiquitin-protein isopeptide ligase (E3) Smurf1 induces Runx2 degradation (220) and Smad6 enhances Smurf1-induced Runx2 degradation (221). For example, TNFa attenuates osteoblast differentiation by promoting Runx2 proteasomal degradation through up-regulation of Smurf1 and Smurf2 expression (222). Transgenic overexpression of Smurf1 in murine osteoblasts suppresses their differentiation and bone formation, while Smurf1-deficient mice develop age-dependent increases in bone mass (111, 223). HDAC4 and HDAC5 deacetylate Runx2, allowing the protein to undergo Smurf-mediated degradation (215).
6.1.3 Transcription factor partners of Runx2
Many transcription factors involved in regulation of the osteoblast differentiation process exert their action by interacting with Runx2 (Figure 2). Some provide costimulatory signals, while others directly repress Runx2 function by affecting its DNA binding activity and/or transactivation potential. DNA binding proteins that interact and cooperate with Runx2 to activate gene expression include AP1 (c-Fos and c-Jun)(224), BMP-responsive Smads (Smad1 and Smad5)(94, 225, 226), Ets1 (227), C/EBPß and -d (228, 229), Dlx5 (230, 231), Hes1 (232), and Menin (233) (Table 1). Most of these proteins interact with either the DNA binding domain or the activation domain of Runx2, although the binding sites for some have not been defined. It is generally believed that these transcription factors cooperate with Runx2 to facilitate the recruitment of coactivators and the assembly of higher-order transactivation complexes. Some proteins, including Hes1, may perturb TLE-Runx interaction both by competing with TLE corepressors for the binding site on Runx2 and by titrating TLE away from Runx2 (232). Furthermore, there is increasing evidence that Dlx5 promotes activation of Osteocalcin by forming heterodimers with Msx2, which antagonizes the Msx2-mediated repression of Osteocalcin (230, 234). Other cooperating transcription factors, such as AP1, Smad1, and Smad5, integrate Runx2 with cell signaling pathways and to the extracellular environment (235, 236). Mutations that affect Runx2-Smad interactions are found in CCD patients and inhibit the ability of Runx2 to induce osteoblast differentiation after BMP stimulation (90).
Transcription factors that inhibit Runx2-dependent activation in mesenchymal cells or osteoblasts include Dlx3 (231), Lef1 (237), Msx2 (230), PPAR? (238), Smad3 (239), Hey1 (240), and Stat1 (241). These proteins repress Runx2 via several mechanisms, including binding the Runt domain and preventing DNA binding (e.g., Lef1, PPAR?) (237, 238), sequestering Runx2 in the cytoplasm (e.g., C/EBPd, Stat1) (241, 242), or unknown mechanisms that involve binding in or around the nuclear matrix targeting domain of Runx2 (e.g., Dlx3) (231).
6.2 Osterix
The discovery of a BMP2-inducible gene, Osx, a Kruppel-like Sp-1 binding factor, identified a second transcriptional regulator for the final stages of bone tissue formation (7). Osx contains a DNA-binding domain consisting of three C2H2-type zinc fingers at its C terminus that share a high degree of sequence identity with similar motifs in Spl, Sp3, and Sp4. In addition, Osx also contains a proline- and serine-rich transactivation domain and activates the OCN and Col1a1 genes. In Osx-null mutant mice, no endochondral or intramembranous bone formation occurs (Table 1) (7). The mesenchymal cells in Osx-null mutant mice do not deposit bone matrix, and cells in the periosteum and the condensed mesenchyme of membranous skeletal elements cannot differentiate into osteoblasts, although these cells express normal levels of Runx2. Interestingly, Osx-null osteoblast precursors in the periosteum of membranous bones express chondrocyte markers, such as Sox9 and Col2a1, suggesting that Runx2-expressing preosteoblasts are still bipotential cells and Osx acts downstream of Runx2 to induce osteoblastic differentiation in bipotential osteochondroprogenitor cells (Figure 2) (7). Currently, there is no evidence to indicate whether Runx2 and Osx functionally or physically interact. Milona et al. have reported that there is an OSE2 element in the Specificity protein-7 (Sp7; the human homologue of the mouse Osterix gene) regulatory region, so the Osx promoter may be a direct target of Runx2 (243). Further analysis on the relationship between Runx2 and Osx will be the focus of future studies.
Several studies implicate the presence of Runx2-independent mechanisms for ossification (60, 185). These studies implicate that additional signaling pathways may act in parallel to, or independent of, Runx2 during osteoblast differentiation. It has been shown that MAPK and PKD signaling pathways serve as points of convergence for mediating the BMP-2- and IGF-I-induced effects on Osx expression in mesenchymal stem cells (244). Additionally, Runx2 was required but not sufficient for the BMP-2-mediated Osx induction. This result indicated that additional factors (e.g. Dlx5) acting downstream of BMP-2 could induce Osx independent of the levels of Runx2 activity. Mechanistic analyses of the effect of FK506 on bone mass showed that NFAT cooperates with Osterix and accelerates osteoblast differentiation and bone formation(245). Overexpression of NFATc1 stimulates Osterix-dependent activation of the Col1a1 promoter, but not Runx2-dependent activation of the Bglap1 (encoding osteocalcin) promoter.
En1 expression temporally precedes that of the osteogenic determinant Osx, and, in the absence of En1, the onset of Osx expression is delayed. As Osx is necessary for potentiating the osteogenic fate of the skeletogenic mesenchyme (7), its perturbed expression provides a mechanistic basis for the delayed calvarial ossification in En1−/− mice (246). Furthermore, that Osx expression remains impaired in En1-null osteoblasts, suggests that En1 also lies upstream of Osx during later phases of calvarial osteogenesis, and thus mediates distinct functions in osteoblast differentiation. En1−/− osteoblasts were deficient in mediating osteoid mineralization and exhibited reduced ALP activity, an enzyme that is essential for this process (8, 247). Moreover, ablation of En1 results in impaired Ocn and Bsp expression, genes that are normally associated with advanced osteoblast differentiation. Ocn expression has also been shown to be dependent on Osx (7).
6.3 ATF4
Using a combination of human genetic information, analysis of mutant mouse strains, and molecular studies, Yang et al. identify activating transcription factor (ATF4) as a critical substrate of RSK2 that is required for the timely onset of osteoblast differentiation, for terminal differentiation of osteoblasts, and for Bsp, and Osteocalcin expression (Figure 2) (248). Additionally, RSK2 and ATF4 posttranscriptionally regulate the synthesis of type I collagen, the main constituent of the bone matrix. These findings identify ATF4 as a critical regulator of osteoblast differentiation and function, and indicate that lack of ATF4 phosphorylation by RSK2 may contribute to the skeletal phenotype of Coffin-Lowry Syndrome (CLS) (Table 1) (248). In addition, treatment of non-osteoblastic cells with MG115, a proteasome inhibitor, induced ATF4 accumulation and resulted in activation of an Osteocalcin promoter luciferase construct as well as expression of endogenous Osteocalcin, a molecular marker of differentiated osteoblasts (249). This study establishes that ATF4, like other osteoblast differentiation factors, such as Runx2 and Osterix, has the ability to induce osteoblast-specific gene expression in non-osteoblastic cells.
Cooperative interactions between activating transcription factor 4 and Runx2/Cbfa1 stimulate osteoblast-specific osteocalcin gene expression (250).
6.4 SATB2
The patterning of skeletal elements and bone formation are generally thought to represent distinct pathways; however, evidence is emerging for crosstalk between these processes. This is illustrated in studies that establish the functional role of Hoxa2 in skeletal development, an inhibitor of bone formation and regulator of branchial arch patterning (188, 251). Understanding the mechanisms that mediate these dual roles of Hoxa2 will provide valuable insight into coordination of pathways governing bone patterning and differentiation (252). Grosschedl and colleagues make an important stride toward this goal by demonstrating that the nuclear matrix protein Satb2 represses Hoxa2 expression and is an activator of multiple steps of Runx2-dependent osteoblast differentiation (Figure 2) (253).
SATB2 is a recently cloned member of the family of special AT-rich binding proteins that binds to nuclear matrix-attachment regions (MARs) and activates transcription in a MAR-dependent manner (254, 255). In humans, translocations that involve the chromosomal region 2q32–q33 and are associated with a cleft palate under conditions of haploinsufficiency have been found to interrupt the SATB2 gene (256). Using targeted mutagenesis of Satb2 in mice, Dobreva et al. provide insight into how the nuclear matrix, chromatin remodeling, and gene activation come together to regulate osteoblast differentiation in development (253). The most striking phenotypes detected in mice lacking Satb2 are craniofacial defects in skeletal elements and the inhibition of normal osteoblast differentiation (Table 1). By combining an impressive series of molecular and genetic approaches, the authors reveal that SATB2 represses Hoxa2 expression in osteoblasts through direct recognition of a MAR-like sequence (Figure 2). Chromatin immunoprecipitation (ChIP) and transactivation experiments reveal that Satb2 can bind to and regulate Bsp and Ocn genes, themselves critical components in osteoblast formation. This strongly suggests that Satb2 has multiple inputs into transcriptional control of osteoblast differentiation (Figure 2). On the basis of genetic synergy between mouse mutants, protein interaction and transactivation analyses, Dobreva et al. discovered that SATB2 directly interacts with and enhances the activity of both Runx2 and ATF4 (253). The interaction of SATB2 with ATF4 and Runx2 augment their binding to the cognate DNA-recognition elements, although SATB2 does not bind itself to the OSE1 and OSE2 sequences. SATB2 has been shown to recognize specific sequences, termed MAR sequences (254), and indeed, SATB2 was found to regulate the Bsp promoter by binding to a site that resembles a bona fide MAR element. Therefore, SATB2 can act not only as an activating or repressing DNA bound protein but also as a protein scaffold that enhances the activity of other DNA binding proteins. By its ability to regulate the expression or activity of multiple key determinants of skeletal development, SATB2 appears to represent a molecular node of a transcriptional network underlying this process (253). Distinguishing between the different modes of Satb2 activity will be important for a detailed understanding of how the nuclear matrix, chromatin structure, and transcriptional activity coordinate the regulation of multiple steps during osteoblast differentiation (252).
7. SUMMARY AND FUTURE DIRECTIONS
Formation of skeletal elements during embryogenesis and the dynamic remodeling of bone in the adult involve an exquisite interplay of developmental cues, signaling proteins, transcription factors, and their coregulatory proteins that support differentiation of osteogenic lineage cells from the initial mesenchymal progenitor cell to the mature osteocyte in mineralized connective tissue (168). The canonical pathway is an enticing target for developing drugs to battle skeletal diseases as Wnt/ß-catenin signaling is composed of a series of molecular interactions that offer potential places for pharmacological intervention (77). Several unresolved issues that continue to perplex us are offered here in the hope that this will stimulate their resolution. There is considerable evidence to show that Wnt signaling can cooperate in unexpected ways with other pathways. The functional interaction of Wnt signaling with other pathways probably accounts for the observation that Wnt/ß-catenin signaling regulates distinct sets of genes in different cells. The mechanisms by which this combinatorial signaling occurs, and whether it is functionally significant in the diseases and potential therapies, are largely unknown.
Although we have learned much in recent years regarding the role of canonical Wnt signaling in bone formation, some important questions remain to be addressed. For example, we know that most Wnts and FZDs are expressed in bone, but is there a role for Wnt and FZD specificity in the control of osteoblast physiology? The canonical pathway is clearly important for the regulation of bone formation, but do noncanonical pathways also play a role in bone metabolism? Finally, canonical Wnt signaling appears to control osteoclastogenesis through actions on osteoblasts, but do Wnts also have direct effects on bone resorbing cells? These and other questions are likely to be answered in the coming years.
Runx2 is essential for mesenchymal condensation, osteoblast development, and osteoblast maturation. Like all developmentally important proteins, multiple mechanisms actively control Runx2 activity. Since it is currently unknown how Runx2 is regulated during the development of the skeletal system, further studies are required to clarify the detailed mechanism of the temporal and spatial regulation of Runx2 for the normal development and homeostatic regulation of the skeletal system. Additional proteins will be added to the list of Runx2-interacting proteins in the next decade. Mechanisms regulating expression and subsequent activity of Osx, ATF, and SATB2 at different stages of osteoblast maturation are only beginning to be understood. Developmental regulation of ATF and SATB2 and cofactor interactions, as well as the possibility that interactions, especially those with other transcription factors, may be either stimulatory or inhibitory depending on the promoter/enhancer context, are important considerations. Finally, insights into the mechanisms by which proteins and developmental signals affect these transcription factors' subcellular localization to the nuclear matrix will be needed to understand how they regulate gene expression.
ACKNOWLEDGMENTS
We would like to thank Ms. Carrie Soltanoff for assistance with the manuscript. We apologize to the many researchers whose work could not be cited due to space limitations. Work in our laboratory is supported by grants by NIH grant AR44741 (Y.-P. Li) and AR-48133 (Y.-P. Li).
9. REFERENCES
- 1.Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J Clin Invest. 2005;115:3318–25. doi: 10.1172/JCI27071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Stokstad E. Bone quality fills holes in fracture risk. Science. 2005;308:1580. doi: 10.1126/science.308.5728.1580. [DOI] [PubMed] [Google Scholar]
- 3.Jackson ME, Shalhoub V, Lian JB, Stein GS, Marks SC., Jr. Aberrant gene expression in cultured mammalian bone cells demonstrates an osteoblast defect in osteopetrosis. J Cell Biochem. 1994;55:366–72. doi: 10.1002/jcb.240550314. [DOI] [PubMed] [Google Scholar]
- 4.Shalhoub V, Bettencourt B, Jackson ME, MacKay CA, Glimcher MJ, Marks SC, Jr., Stein GS, Lian JB. Abnormalities of phosphoprotein gene expression in three osteopetrotic rat mutations: elevated mRNA transcripts, protein synthesis, and accumulation in bone of mutant animals. J Cell Physiol. 1994;158:110–20. doi: 10.1002/jcp.1041580114. [DOI] [PubMed] [Google Scholar]
- 5.Shalhoub V, Jackson ME, Lian JB, Stein GS, Marks SC., Jr. Gene expression during skeletal development in three osteopetrotic rat mutations. Evidence for osteoblast abnormalities. J Biol Chem. 1991;266:9847–56. [PubMed] [Google Scholar]
- 6.Ducy P, Karsenty G. Two distinct osteoblast-specific cis-acting elements control expression of a mouse osteocalcin gene. Mol Cell Biol. 1995;15:1858–69. doi: 10.1128/mcb.15.4.1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, de Crombrugghe B. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 2002;108:17–29. doi: 10.1016/s0092-8674(01)00622-5. [DOI] [PubMed] [Google Scholar]
- 8.Murshed M, Harmey D, Millan JL, McKee MD, Karsenty G. Unique coexpression in osteoblasts of broadly expressed genes accounts for the spatial restriction of ECM mineralization to bone. Genes Dev. 2005;19:1093–104. doi: 10.1101/gad.1276205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Schweitzer MH, Wittmeyer JL, Horner JR, Toporski JK. Soft-tissue vessels and cellular preservation in Tyrannosaurus rex. Science. 2005;307:1952–5. doi: 10.1126/science.1108397. [DOI] [PubMed] [Google Scholar]
- 10.Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, Manolagas SC, Bellido T. Prevention of osteocyte and osteoblast apoptosis by bisphosphonates and calcitonin. J Clin Invest. 1999;104:1363–74. doi: 10.1172/JCI6800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Narayanan K, Ramachandran A, Hao J, He G, Park KW, Cho M, George A. Dual functional roles of dentin matrix protein 1. Implications in biomineralization and gene transcription by activation of intracellular Ca2+ store. J Biol Chem. 2003;278:17500–8. doi: 10.1074/jbc.M212700200. [DOI] [PubMed] [Google Scholar]
- 12.Ye L, Mishina Y, Chen D, Huang H, Dallas SL, Dallas MR, Sivakumar P, Kunieda T, Tsutsui TW, Boskey A, Bonewald LF, Feng JQ. Dmp1-deficient mice display severe defects in cartilage formation responsible for a chondrodysplasia-like phenotype. J Biol Chem. 2005;280:6197–203. doi: 10.1074/jbc.M412911200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ling Y, Rios HF, Myers ER, Lu Y, Feng JQ, Boskey AL. DMP1 depletion decreases bone mineralization in vivo: an FTIR imaging analysis. J Bone Miner Res. 2005;20:2169–77. doi: 10.1359/JBMR.050815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lorenz-Depiereux B, Bastepe M, Benet-Pages A, Amyere M, Wagenstaller J, Muller-Barth U, Badenhoop K, Kaiser SM, Rittmaster RS, Shlossberg AH, Olivares JL, Loris C, Ramos FJ, Glorieux F, Vikkula M, Juppner H, Strom TM. DMP1 mutations in autosomal recessive hypophosphatemia implicate a bone matrix protein in the regulation of phosphate homeostasis. Nat Genet. 2006 doi: 10.1038/ng1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Karsenty G, Wagner EF. Reaching a genetic and molecular understanding of skeletal development. Dev Cell. 2002;2:389–406. doi: 10.1016/s1534-5807(02)00157-0. [DOI] [PubMed] [Google Scholar]
- 16.Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, Kondo H, Richards WG, Bannon TW, Noda M, Clement K, Vaisse C, Karsenty G. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature. 2005;434:514–20. doi: 10.1038/nature03398. [DOI] [PubMed] [Google Scholar]
- 17.Winslow MM, Pan M, Starbuck M, Gallo EM, Deng L, Karsenty G, Crabtree GR. Calcineurin/NFAT signaling in osteoblasts regulates bone mass. Dev Cell. 2006;10:771–82. doi: 10.1016/j.devcel.2006.04.006. [DOI] [PubMed] [Google Scholar]
- 18.Taichman RS. Blood and bone: two tissues whose fates are intertwined to create the hematopoietic stem-cell niche. Blood. 2005;105:2631–9. doi: 10.1182/blood-2004-06-2480. [DOI] [PubMed] [Google Scholar]
- 19.Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, Harris S, Wiedemann LM, Mishina Y, Li L. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425:836–41. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
- 20.Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–6. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
- 21.Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006;311:1880–5. doi: 10.1126/science.1110542. [DOI] [PubMed] [Google Scholar]
- 22.Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, Ito K, Koh GY, Suda T. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118:149–61. doi: 10.1016/j.cell.2004.07.004. [DOI] [PubMed] [Google Scholar]
- 23.Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121:1109–21. doi: 10.1016/j.cell.2005.05.026. [DOI] [PubMed] [Google Scholar]
- 24.Papayannopoulou T. Current mechanistic scenarios in hematopoietic stem/progenitor cell mobilization. Blood. 2004;103:1580–5. doi: 10.1182/blood-2003-05-1595. [DOI] [PubMed] [Google Scholar]
- 25.Levesque JP, Liu F, Simmons PJ, Betsuyaku T, Senior RM, Pham C, Link DC. Characterization of hematopoietic progenitor mobilization in protease-deficient mice. Blood. 2004;104:65–72. doi: 10.1182/blood-2003-05-1589. [DOI] [PubMed] [Google Scholar]
- 26.Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, Frenette PS. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407–21. doi: 10.1016/j.cell.2005.10.041. [DOI] [PubMed] [Google Scholar]
- 27.Larsson J, Scadden D. Nervous activity in a stem cell niche. Cell. 2006;124:253–5. doi: 10.1016/j.cell.2006.01.006. [DOI] [PubMed] [Google Scholar]
- 28.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
- 29.Watt FM. Stem cell fate and patterning in mammalian epidermis. Curr Opin Genet Dev. 2001;11:410–7. doi: 10.1016/s0959-437x(00)00211-2. [DOI] [PubMed] [Google Scholar]
- 30.Gronthos S, Zannettino AC, Graves SE, Ohta S, Hay SJ, Simmons PJ. Differential cell surface expression of the STRO-1 and alkaline phosphatase antigens on discrete developmental stages in primary cultures of human bone cells. J Bone Miner Res. 1999;14:47–56. doi: 10.1359/jbmr.1999.14.1.47. [DOI] [PubMed] [Google Scholar]
- 31.Stein GS, Lian JB, Stein JL, Van Wijnen AJ, Montecino M. Transcriptional control of osteoblast growth and differentiation. Physiol Rev. 1996;76:593–629. doi: 10.1152/physrev.1996.76.2.593. [DOI] [PubMed] [Google Scholar]
- 32.Tamai K, Semenov M, Kato Y, Spokony R, Liu C, Katsuyama Y, Hess F, Saint-Jeannet JP, He X. LDL-receptor-related proteins in Wnt signal transduction. Nature. 2000;407:530–5. doi: 10.1038/35035117. [DOI] [PubMed] [Google Scholar]
- 33.He X, Semenov M, Tamai K, Zeng X. LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way. Development. 2004;131:1663–77. doi: 10.1242/dev.01117. [DOI] [PubMed] [Google Scholar]
- 34.Veeman MT, Axelrod JD, Moon RT. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell. 2003;5:367–77. doi: 10.1016/s1534-5807(03)00266-1. [DOI] [PubMed] [Google Scholar]
- 35.Westendorf JJ, Kahler RA, Schroeder TM. Wnt signaling in osteoblasts and bone diseases. Gene. 2004;341:19–39. doi: 10.1016/j.gene.2004.06.044. [DOI] [PubMed] [Google Scholar]
- 36.Rawadi G, Roman-Roman S. Wnt signalling pathway: a new target for the treatment of osteoporosis. Expert Opin Ther Targets. 2005;9:1063–77. doi: 10.1517/14728222.9.5.1063. [DOI] [PubMed] [Google Scholar]
- 37.Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781–810. doi: 10.1146/annurev.cellbio.20.010403.113126. [DOI] [PubMed] [Google Scholar]
- 38.Moon RT, Kohn AD, De Ferrari GV, Kaykas A. WNT and beta-catenin signalling: diseases and therapies. Nat Rev Genet. 2004;5:691–701. doi: 10.1038/nrg1427. [DOI] [PubMed] [Google Scholar]
- 39.Hay E, Faucheu C, Suc-Royer I, Touitou R, Stiot V, Vayssiere B, Baron R, Roman-Roman S, Rawadi G. Interaction between LRP5 and Frat1 mediates the activation of the Wnt canonical pathway. J Biol Chem. 2005;280:13616–23. doi: 10.1074/jbc.M411999200. [DOI] [PubMed] [Google Scholar]
- 40.Clevers H. Wnt signaling: Ig-norrin the dogma. Curr Biol. 2004;14:R436–7. doi: 10.1016/j.cub.2004.05.039. [DOI] [PubMed] [Google Scholar]
- 41.Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 2004;303:1483–7. doi: 10.1126/science.1094291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Levy L, Wei Y, Labalette C, Wu Y, Renard CA, Buendia MA, Neuveut C. Acetylation of beta-catenin by p300 regulates beta-catenin-Tcf4 interaction. Mol Cell Biol. 2004;24:3404–14. doi: 10.1128/MCB.24.8.3404-3414.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Semenov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem. 2005;280:26770–5. doi: 10.1074/jbc.M504308200. [DOI] [PubMed] [Google Scholar]
- 44.Mao B, Wu W, Davidson G, Marhold J, Li M, Mechler BM, Delius H, Hoppe D, Stannek P, Walter C, Glinka A, Niehrs C. Kremen proteins are Dickkopf receptors that regulate Wnt/beta-catenin signalling. Nature. 2002;417:664–7. doi: 10.1038/nature756. [DOI] [PubMed] [Google Scholar]
- 45.Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A, Niehrs C. LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature. 2001;411:321–5. doi: 10.1038/35077108. [DOI] [PubMed] [Google Scholar]
- 46.Li X, Liu P, Liu W, Maye P, Zhang J, Zhang Y, Hurley M, Guo C, Boskey A, Sun L, Harris SE, Rowe DW, Ke HZ, Wu D. Dkk2 has a role in terminal osteoblast differentiation and mineralized matrix formation. Nat Genet. 2005;37:945–52. doi: 10.1038/ng1614. [DOI] [PubMed] [Google Scholar]
- 47.Kieslinger M, Folberth S, Dobreva G, Dorn T, Croci L, Erben R, Consalez GG, Grosschedl R. EBF2 regulates osteoblast-dependent differentiation of osteoclasts. Dev Cell. 2005;9:757–67. doi: 10.1016/j.devcel.2005.10.009. [DOI] [PubMed] [Google Scholar]
- 48.Tan C, Costello P, Sanghera J, Dominguez D, Baulida J, de Herreros AG, Dedhar S. Inhibition of integrin linked kinase (ILK) suppresses beta-catenin-Lef/Tcf-dependent transcription and expression of the E-cadherin repressor, snail, in APC−/− human colon carcinoma cells. Oncogene. 2001;20:133–40. doi: 10.1038/sj.onc.1204052. [DOI] [PubMed] [Google Scholar]
- 49.Levina E, Oren M, Ben-Ze'ev A. Downregulation of beta-catenin by p53 involves changes in the rate of beta-catenin phosphorylation and Axin dynamics. Oncogene. 2004;23:4444–53. doi: 10.1038/sj.onc.1207587. [DOI] [PubMed] [Google Scholar]
- 50.Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci. 2003;116:1175–86. doi: 10.1242/jcs.00384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Moon RT, Bowerman B, Boutros M, Perrimon N. The promise and perils of Wnt signaling through beta-catenin. Science. 2002;296:1644–6. doi: 10.1126/science.1071549. [DOI] [PubMed] [Google Scholar]
- 52.Mbalaviele G, Sheikh S, Stains JP, Salazar VS, Cheng SL, Chen D, Civitelli R. Beta-catenin and BMP-2 synergize to promote osteoblast differentiation and new bone formation. J Cell Biochem. 2005;94:403–18. doi: 10.1002/jcb.20253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Abe E, Marians RC, Yu W, Wu XB, Ando T, Li Y, Iqbal J, Eldeiry L, Rajendren G, Blair HC, Davies TF, Zaidi M. TSH is a negative regulator of skeletal remodeling. Cell. 2003;115:151–62. doi: 10.1016/s0092-8674(03)00771-2. [DOI] [PubMed] [Google Scholar]
- 54.Yamaguchi TP, Bradley A, McMahon AP, Jones S. A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development. 1999;126:1211–23. doi: 10.1242/dev.126.6.1211. [DOI] [PubMed] [Google Scholar]
- 55.Yang Y, Topol L, Lee H, Wu J. Wnt5a and Wnt5b exhibit distinct activities in coordinating chondrocyte proliferation and differentiation. Development. 2003;130:1003–15. doi: 10.1242/dev.00324. [DOI] [PubMed] [Google Scholar]
- 56.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, Borrone C, Brunner HG, Carle GF, Dallapiccola B, De Paepe A, Floege B, Halfhide ML, Hall B, Hennekam RC, Hirose T, Jans A, Juppner H, Kim CA, Keppler-Noreuil K, Kohlschuetter A, LaCombe D, Lambert M, Lemyre E, Letteboer T, Peltonen L, Ramesar RS, Romanengo M, Somer H, Steichen-Gersdorf E, Steinmann B, Sullivan B, Superti-Furga A, Swoboda W, van den Boogaard MJ, Van Hul W, Vikkula M, Votruba M, Zabel B, Garcia T, Baron R, Olsen BR, Warman ML. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513–23. doi: 10.1016/s0092-8674(01)00571-2. [DOI] [PubMed] [Google Scholar]
- 57.Ai M, Holmen SL, Van Hul W, Williams BO, Warman ML. Reduced affinity to and inhibition by DKK1 form a common mechanism by which high bone mass-associated missense mutations in LRP5 affect canonical Wnt signaling. Mol Cell Biol. 2005;25:4946–55. doi: 10.1128/MCB.25.12.4946-4955.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick MA, Wu D, Insogna K, Lifton RP. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med. 2002;346:1513–21. doi: 10.1056/NEJMoa013444. [DOI] [PubMed] [Google Scholar]
- 59.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. 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. 2003;72:763–71. doi: 10.1086/368277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass DA, 2nd, Hartmann C, Li L, Hwang TH, Brayton CF, Lang RA, Karsenty G, Chan L. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–14. doi: 10.1083/jcb.200201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Babij P, Zhao W, Small C, Kharode Y, Yaworsky PJ, Bouxsein ML, Reddy PS, Bodine PV, Robinson JA, Bhat B, Marzolf J, Moran RA, Bex F. High bone mass in mice expressing a mutant LRP5 gene. J Bone Miner Res. 2003;18:960–74. doi: 10.1359/jbmr.2003.18.6.960. [DOI] [PubMed] [Google Scholar]
- 62.Bain G, Muller T, Wang X, Papkoff J. Activated beta-catenin induces osteoblast differentiation of C3H10T1/2 cells and participates in BMP2 mediated signal transduction. Biochem Biophys Res Commun. 2003;301:84–91. doi: 10.1016/s0006-291x(02)02951-0. [DOI] [PubMed] [Google Scholar]
- 63.Rawadi G, Vayssiere B, Dunn F, Baron R, Roman-Roman S. BMP-2 controls alkaline phosphatase expression and osteoblast mineralization by a Wnt autocrine loop. J Bone Miner Res. 2003;18:1842–53. doi: 10.1359/jbmr.2003.18.10.1842. [DOI] [PubMed] [Google Scholar]
- 64.Bennett CN, Longo KA, Wright WS, Suva LJ, Lane TF, Hankenson KD, MacDougald OA. Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc Natl Acad Sci U S A. 2005;102:3324–9. doi: 10.1073/pnas.0408742102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jackson A, Vayssiere B, Garcia T, Newell W, Baron R, Roman-Roman S, Rawadi G. Gene array analysis of Wnt-regulated genes in C3H10T1/2 cells. Bone. 2005;36:585–98. doi: 10.1016/j.bone.2005.01.007. [DOI] [PubMed] [Google Scholar]
- 66.Stambolic V, Ruel L, Woodgett JR. Lithium inhibits glycogen synthase kinase-3 activity and mimics wingless signalling in intact cells. Curr Biol. 1996;6:1664–8. doi: 10.1016/s0960-9822(02)70790-2. [DOI] [PubMed] [Google Scholar]
- 67.Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005;8:739–50. doi: 10.1016/j.devcel.2005.03.016. [DOI] [PubMed] [Google Scholar]
- 68.Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. Canonical Wnt/beta-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell. 2005;8:727–38. doi: 10.1016/j.devcel.2005.02.013. [DOI] [PubMed] [Google Scholar]
- 69.Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, Deng L, Clemens TL, Williams BO. Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 2005;280:21162–8. doi: 10.1074/jbc.M501900200. [DOI] [PubMed] [Google Scholar]
- 70.Clement-Lacroix P, Ai M, Morvan F, Roman-Roman S, Vayssiere B, Belleville C, Estrera K, Warman ML, Baron R, Rawadi G. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci U S A. 2005;102:17406–11. doi: 10.1073/pnas.0505259102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Boland GM, Perkins G, Hall DJ, Tuan RS. Wnt 3a promotes proliferation and suppresses osteogenic differentiation of adult human mesenchymal stem cells. J Cell Biochem. 2004;93:1210–30. doi: 10.1002/jcb.20284. [DOI] [PubMed] [Google Scholar]
- 72.de Boer J, Siddappa R, Gaspar C, van Apeldoorn A, Fodde R, van Blitterswijk C. Wnt signaling inhibits osteogenic differentiation of human mesenchymal stem cells. Bone. 2004;34:818–26. doi: 10.1016/j.bone.2004.01.016. [DOI] [PubMed] [Google Scholar]
- 73.Luo Q, Kang Q, Si W, Jiang W, Park JK, Peng Y, Li X, Luu HH, Luo J, Montag AG, Haydon RC, He TC. Connective tissue growth factor (CTGF) is regulated by Wnt and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells. J Biol Chem. 2004;279:55958–68. doi: 10.1074/jbc.M407810200. [DOI] [PubMed] [Google Scholar]
- 74.Glass DA, 2nd, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, McMahon AP, Lang RA, Karsenty G. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005;8:751–64. doi: 10.1016/j.devcel.2005.02.017. [DOI] [PubMed] [Google Scholar]
- 75.Hu H, Hilton MJ, Tu X, Yu K, Ornitz DM, Long F. Sequential roles of Hedgehog and Wnt signaling in osteoblast development. Development. 2005;132:49–60. doi: 10.1242/dev.01564. [DOI] [PubMed] [Google Scholar]
- 76.Tao Q, Yokota C, Puck H, Kofron M, Birsoy B, Yan D, Asashima M, Wylie CC, Lin X, Heasman J. Maternal wnt11 activates the canonical wnt signaling pathway required for axis formation in Xenopus embryos. Cell. 2005;120:857–71. doi: 10.1016/j.cell.2005.01.013. [DOI] [PubMed] [Google Scholar]
- 77.Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt signaling. J Clin Invest. 2006;116:1202–9. doi: 10.1172/JCI28551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gaur T, Lengner CJ, Hovhannisyan H, Bhat RA, Bodine PV, Komm BS, Javed A, van Wijnen AJ, Stein JL, Stein GS, Lian JB. Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 2005;280:33132–40. doi: 10.1074/jbc.M500608200. [DOI] [PubMed] [Google Scholar]
- 79.Henriksen K, Gram J, Hoegh-Andersen P, Jemtland R, Ueland T, Dziegiel MH, Schaller S, Bollerslev J, Karsdal MA. Osteoclasts from patients with autosomal dominant osteopetrosis type I caused by a T253I mutation in low-density lipoprotein receptor-related protein 5 are normal in vitro, but have decreased resorption capacity in vivo. Am J Pathol. 2005;167:1341–8. doi: 10.1016/S0002-9440(10)61221-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Canalis E, Economides AN, Gazzerro E. Bone morphogenetic proteins, their antagonists, and the skeleton. Endocr Rev. 2003;24:218–35. doi: 10.1210/er.2002-0023. [DOI] [PubMed] [Google Scholar]
- 81.Chen D, Zhao M, Mundy GR. Bone morphogenetic proteins. Growth Factors. 2004;22:233–41. doi: 10.1080/08977190412331279890. [DOI] [PubMed] [Google Scholar]
- 82.Cao X, Chen D. The BMP signaling and in vivo bone formation. Gene. 2005;357:1–8. doi: 10.1016/j.gene.2005.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Anderson HC, Hodges PT, Aguilera XM, Missana L, Moylan PE. Bone morphogenetic protein (BMP) localization in developing human and rat growth plate, metaphysis, epiphysis, and articular cartilage. J Histochem Cytochem. 2000;48:1493–502. doi: 10.1177/002215540004801106. [DOI] [PubMed] [Google Scholar]
- 84.Hoffmann A, Gross G. BMP signaling pathways in cartilage and bone formation. Crit Rev Eukaryot Gene Expr. 2001;11:23–45. [PubMed] [Google Scholar]
- 85.Peng Y, Kang Q, Cheng H, Li X, Sun MH, Jiang W, Luu HH, Park JY, Haydon RC, He TC. Transcriptional characterization of bone morphogenetic proteins (BMPs)-mediated osteogenic signaling. J Cell Biochem. 2003;90:1149–65. doi: 10.1002/jcb.10744. [DOI] [PubMed] [Google Scholar]
- 86.Daluiski A, Engstrand T, Bahamonde ME, Gamer LW, Agius E, Stevenson SL, Cox K, Rosen V, Lyons KM. Bone morphogenetic protein-3 is a negative regulator of bone density. Nat Genet. 2001;27:84–8. doi: 10.1038/83810. [DOI] [PubMed] [Google Scholar]
- 87.Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001;29:117–29. doi: 10.1038/ng1001-117. [DOI] [PubMed] [Google Scholar]
- 88.Miyazono K, Maeda S, Imamura T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev. 2005;16:251–63. doi: 10.1016/j.cytogfr.2005.01.009. [DOI] [PubMed] [Google Scholar]
- 89.Massague J. How cells read TGF-beta signals. Nat Rev Mol Cell Biol. 2000;1:169–78. doi: 10.1038/35043051. [DOI] [PubMed] [Google Scholar]
- 90.Zhang YW, Yasui N, Ito K, Huang G, Fujii M, Hanai J, Nogami H, Ochi T, Miyazono K, Ito Y. A RUNX2/PEBP2alpha A/CBFA1 mutation displaying impaired transactivation and Smad interaction in cleidocranial dysplasia. Proc Natl Acad Sci U S A. 2000;97:10549–54. doi: 10.1073/pnas.180309597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Miyazono K, Maeda S, Imamura T. Coordinate regulation of cell growth and differentiation by TGF-beta superfamily and Runx proteins. Oncogene. 2004;23:4232–7. doi: 10.1038/sj.onc.1207131. [DOI] [PubMed] [Google Scholar]
- 92.Ito Y, Miyazono K. RUNX transcription factors as key targets of TGF-beta superfamily signaling. Curr Opin Genet Dev. 2003;13:43–7. doi: 10.1016/s0959-437x(03)00007-8. [DOI] [PubMed] [Google Scholar]
- 93.Maeda S, Hayashi M, Komiya S, Imamura T, Miyazono K. Endogenous TGF-beta signaling suppresses maturation of osteoblastic mesenchymal cells. Embo J. 2004;23:552–63. doi: 10.1038/sj.emboj.7600067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lee KS, Kim HJ, Li QL, Chi XZ, Ueta C, Komori T, Wozney JM, Kim EG, Choi JY, Ryoo HM, Bae SC. Runx2 is a common target of transforming growth factor beta1 and bone morphogenetic protein 2, and cooperation between Runx2 and Smad5 induces osteoblast-specific gene expression in the pluripotent mesenchymal precursor cell line C2C12. Mol Cell Biol. 2000;20:8783–92. doi: 10.1128/mcb.20.23.8783-8792.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Miyama K, Yamada G, Yamamoto TS, Takagi C, Miyado K, Sakai M, Ueno N, Shibuya H. A BMP-inducible gene, dlx5, regulates osteoblast differentiation and mesoderm induction. Dev Biol. 1999;208:123–33. doi: 10.1006/dbio.1998.9197. [DOI] [PubMed] [Google Scholar]
- 96.Lee MH, Kim YJ, Kim HJ, Park HD, Kang AR, Kyung HM, Sung JH, Wozney JM, Kim HJ, Ryoo HM. BMP-2-induced Runx2 expression is mediated by Dlx5, and TGF-beta 1 opposes the BMP-2-induced osteoblast differentiation by suppression of Dlx5 expression. J Biol Chem. 2003;278:34387–94. doi: 10.1074/jbc.M211386200. [DOI] [PubMed] [Google Scholar]
- 97.de Jong DS, Vaes BL, Dechering KJ, Feijen A, Hendriks JM, Wehrens R, Mummery CL, van Zoelen EJ, Olijve W, Steegenga WT. Identification of novel regulators associated with early-phase osteoblast differentiation. J Bone Miner Res. 2004;19:947–58. doi: 10.1359/JBMR.040216. [DOI] [PubMed] [Google Scholar]
- 98.Korchynskyi O, Dechering KJ, Sijbers AM, Olijve W, ten Dijke P. Gene array analysis of bone morphogenetic protein type I receptor-induced osteoblast differentiation. J Bone Miner Res. 2003;18:1177–85. doi: 10.1359/jbmr.2003.18.7.1177. [DOI] [PubMed] [Google Scholar]
- 99.Guicheux J, Lemonnier J, Ghayor C, Suzuki A, Palmer G, Caverzasio J. Activation of p38 mitogen-activated protein kinase and c-Jun-NH2-terminal kinase by BMP-2 and their implication in the stimulation of osteoblastic cell differentiation. J Bone Miner Res. 2003;18:2060–8. doi: 10.1359/jbmr.2003.18.11.2060. [DOI] [PubMed] [Google Scholar]
- 100.Lai CF, Cheng SL. Signal transductions induced by bone morphogenetic protein-2 and transforming growth factor-beta in normal human osteoblastic cells. J Biol Chem. 2002;277:15514–22. doi: 10.1074/jbc.M200794200. [DOI] [PubMed] [Google Scholar]
- 101.Lemonnier J, Ghayor C, Guicheux J, Caverzasio J. Protein kinase C-independent activation of protein kinase D is involved in BMP-2-induced activation of stress mitogen-activated protein kinases JNK and p38 and osteoblastic cell differentiation. J Biol Chem. 2004;279:259–64. doi: 10.1074/jbc.M308665200. [DOI] [PubMed] [Google Scholar]
- 102.Lee KS, Hong SH, Bae SC. Both the Smad and p38 MAPK pathways play a crucial role in Runx2 expression following induction by transforming growth factor-beta and bone morphogenetic protein. Oncogene. 2002;21:7156–63. doi: 10.1038/sj.onc.1205937. [DOI] [PubMed] [Google Scholar]
- 103.Lee MH, Javed A, Kim HJ, Shin HI, Gutierrez S, Choi JY, Rosen V, Stein JL, van Wijnen AJ, Stein GS, Lian JB, Ryoo HM. Transient upregulation of CBFA1 in response to bone morphogenetic protein-2 and transforming growth factor beta1 in C2C12 myogenic cells coincides with suppression of the myogenic phenotype but is not sufficient for osteoblast differentiation. J Cell Biochem. 1999;73:114–25. [PubMed] [Google Scholar]
- 104.Phimphilai M, Zhao Z, Boules H, Roca H, Franceschi RT. BMP signaling is required for RUNX2-dependent induction of the osteoblast phenotype. J Bone Miner Res. 2006;21:637–46. doi: 10.1359/JBMR.060109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yagi K, Tsuji K, Nifuji A, Shinomiya K, Nakashima K, DeCrombrugghe B, Noda M. Bone morphogenetic protein-2 enhances osterix gene expression in chondrocytes. J Cell Biochem. 2003;88:1077–83. doi: 10.1002/jcb.10467. [DOI] [PubMed] [Google Scholar]
- 106.Lee MH, Kwon TG, Park HS, Wozney JM, Ryoo HM. BMP-2-induced Osterix expression is mediated by Dlx5 but is independent of Runx2. Biochem Biophys Res Commun. 2003;309:689–94. doi: 10.1016/j.bbrc.2003.08.058. [DOI] [PubMed] [Google Scholar]
- 107.Celil AB, Hollinger JO, Campbell PG. Osx transcriptional regulation is mediated by additional pathways to BMP2/Smad signaling. J Cell Biochem. 2005;95:518–28. doi: 10.1002/jcb.20429. [DOI] [PubMed] [Google Scholar]
- 108.Izzi L, Attisano L. Regulation of the TGFbeta signalling pathway by ubiquitin-mediated degradation. Oncogene. 2004;23:2071–8. doi: 10.1038/sj.onc.1207412. [DOI] [PubMed] [Google Scholar]
- 109.Datto M, Wang XF. Ubiquitin-mediated degradation a mechanism for fine-tuning TGF-beta signaling. Cell. 2005;121:2–4. doi: 10.1016/j.cell.2005.03.017. [DOI] [PubMed] [Google Scholar]
- 110.Dupont S, Zacchigna L, Cordenonsi M, Soligo S, Adorno M, Rugge M, Piccolo S. Germ-layer specification and control of cell growth by Ectodermin, a Smad4 ubiquitin ligase. Cell. 2005;121:87–99. doi: 10.1016/j.cell.2005.01.033. [DOI] [PubMed] [Google Scholar]
- 111.Yamashita M, Ying SX, Zhang GM, Li C, Cheng SY, Deng CX, Zhang YE. Ubiquitin ligase Smurf1 controls osteoblast activity and bone homeostasis by targeting MEKK2 for degradation. Cell. 2005;121:101–13. doi: 10.1016/j.cell.2005.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Chung UI, Schipani E, McMahon AP, Kronenberg HM. Indian hedgehog couples chondrogenesis to osteogenesis in endochondral bone development. J Clin Invest. 2001;107:295–304. doi: 10.1172/JCI11706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.St-Jacques B, Hammerschmidt M, McMahon AP. Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev. 1999;13:2072–86. doi: 10.1101/gad.13.16.2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Long F, Chung UI, Ohba S, McMahon J, Kronenberg HM, McMahon AP. Ihh signaling is directly required for the osteoblast lineage in the endochondral skeleton. Development. 2004;131:1309–18. doi: 10.1242/dev.01006. [DOI] [PubMed] [Google Scholar]
- 115.Razzaque MS, Soegiarto DW, Chang D, Long F, Lanske B. Conditional deletion of Indian hedgehog from collagen type 2alpha1-expressing cells results in abnormal endochondral bone formation. J Pathol. 2005;207:453–61. doi: 10.1002/path.1870. [DOI] [PubMed] [Google Scholar]
- 116.Nakamura T, Aikawa T, Iwamoto-Enomoto M, Iwamoto M, Higuchi Y, Pacifici M, Kinto N, Yamaguchi A, Noji S, Kurisu K, Matsuya T. Induction of osteogenic differentiation by hedgehog proteins. Biochem Biophys Res Commun. 1997;237:465–9. doi: 10.1006/bbrc.1997.7156. [DOI] [PubMed] [Google Scholar]
- 117.van der Horst G, Farih-Sips H, Lowik CW, Karperien M. Hedgehog stimulates only osteoblastic differentiation of undifferentiated KS483 cells. Bone. 2003;33:899–910. doi: 10.1016/j.bone.2003.07.004. [DOI] [PubMed] [Google Scholar]
- 118.Rodda SJ, McMahon AP. Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development. 2006;133:3231–44. doi: 10.1242/dev.02480. [DOI] [PubMed] [Google Scholar]
- 119.Meng X, Poon R, Zhang X, Cheah A, Ding Q, Hui CC, Alman B. Suppressor of fused negatively regulates beta-catenin signaling. J Biol Chem. 2001;276:40113–9. doi: 10.1074/jbc.M105317200. [DOI] [PubMed] [Google Scholar]
- 120.Jia J, Amanai K, Wang G, Tang J, Wang B, Jiang J. Shaggy/GSK3 antagonizes Hedgehog signalling by regulating Cubitus interruptus. Nature. 2002;416:548–52. doi: 10.1038/nature733. [DOI] [PubMed] [Google Scholar]
- 121.Price MA, Kalderon D. Proteolysis of the Hedgehog signaling effector Cubitus interruptus requires phosphorylation by Glycogen Synthase Kinase 3 and Casein Kinase 1. Cell. 2002;108:823–35. doi: 10.1016/s0092-8674(02)00664-5. [DOI] [PubMed] [Google Scholar]
- 122.Spinella-Jaegle S, Rawadi G, Kawai S, Gallea S, Faucheu C, Mollat P, Courtois B, Bergaud B, Ramez V, Blanchet AM, Adelmant G, Baron R, Roman-Roman S. Sonic hedgehog increases the commitment of pluripotent mesenchymal cells into the osteoblastic lineage and abolishes adipocytic differentiation. J Cell Sci. 2001;114:2085–94. doi: 10.1242/jcs.114.11.2085. [DOI] [PubMed] [Google Scholar]
- 123.Yuasa T, Kataoka H, Kinto N, Iwamoto M, Enomoto-Iwamoto M, Iemura S, Ueno N, Shibata Y, Kurosawa H, Yamaguchi A. Sonic hedgehog is involved in osteoblast differentiation by cooperating with BMP-2. J Cell Physiol. 2002;193:225–32. doi: 10.1002/jcp.10166. [DOI] [PubMed] [Google Scholar]
- 124.Ornitz DM, Marie PJ. FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev. 2002;16:1446–65. doi: 10.1101/gad.990702. [DOI] [PubMed] [Google Scholar]
- 125.Webster MK, Donoghue DJ. Constitutive activation of fibroblast growth factor receptor 3 by the transmembrane domain point mutation found in achondroplasia. Embo J. 1996;15:520–7. [PMC free article] [PubMed] [Google Scholar]
- 126.Wilkie AO. Craniosynostosis: genes and mechanisms. Hum Mol Genet. 1997;6:1647–56. doi: 10.1093/hmg/6.10.1647. [DOI] [PubMed] [Google Scholar]
- 127.Naski MC, Colvin JS, Coffin JD, Ornitz DM. Repression of hedgehog signaling and BMP4 expression in growth plate cartilage by fibroblast growth factor receptor 3. Development. 1998;125:4977–88. doi: 10.1242/dev.125.24.4977. [DOI] [PubMed] [Google Scholar]
- 128.Valverde-Franco G, Liu H, Davidson D, Chai S, Valderrama-Carvajal H, Goltzman D, Ornitz DM, Henderson JE. Defective bone mineralization and osteopenia in young adult FGFR3−/− mice. Hum Mol Genet. 2004;13:271–84. doi: 10.1093/hmg/ddh034. [DOI] [PubMed] [Google Scholar]
- 129.Xiao L, Naganawa T, Obugunde E, Gronowicz G, Ornitz DM, Coffin JD, Hurley MM. Stat1 controls postnatal bone formation by regulating fibroblast growth factor signaling in osteoblasts. J Biol Chem. 2004;279:27743–52. doi: 10.1074/jbc.M314323200. [DOI] [PubMed] [Google Scholar]
- 130.Chen L, Deng CX. Roles of FGF signaling in skeletal development and human genetic diseases. Front Biosci. 2005;10:1961–76. doi: 10.2741/1671. [DOI] [PubMed] [Google Scholar]
- 131.Ornitz DM. FGF signaling in the developing endochondral skeleton. Cytokine Growth Factor Rev. 2005;16:205–13. doi: 10.1016/j.cytogfr.2005.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yamaguchi TP, Harpal K, Henkemeyer M, Rossant J. fgfr-1 is required for embryonic growth and mesodermal patterning during mouse gastrulation. Genes Dev. 1994;8:3032–44. doi: 10.1101/gad.8.24.3032. [DOI] [PubMed] [Google Scholar]
- 133.Li C, Xu X, Nelson DK, Williams T, Kuehn MR, Deng CX. FGFR1 function at the earliest stages of mouse limb development plays an indispensable role in subsequent autopod morphogenesis. Development. 2005;132:4755–64. doi: 10.1242/dev.02065. [DOI] [PubMed] [Google Scholar]
- 134.Partanen J, Schwartz L, Rossant J. Opposite phenotypes of hypomorphic and Y766 phosphorylation site mutations reveal a function for Fgfr1 in anteroposterior patterning of mouse embryos. Genes Dev. 1998;12:2332–44. doi: 10.1101/gad.12.15.2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Verheyden JM, Lewandoski M, Deng C, Harfe BD, Sun X. Conditional inactivation of Fgfr1 in mouse defines its role in limb bud establishment, outgrowth and digit patterning. Development. 2005;132:4235–45. doi: 10.1242/dev.02001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Jacob AL, Smith C, Partanen J, Ornitz DM. Fibroblast growth factor receptor 1 signaling in the osteo-chondrogenic cell lineage regulates sequential steps of osteoblast maturation. Dev Biol. 2006;296:315–28. doi: 10.1016/j.ydbio.2006.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Colvin JS, Feldman B, Nadeau JH, Goldfarb M, Ornitz DM. Genomic organization and embryonic expression of the mouse fibroblast growth factor 9 gene. Dev Dyn. 1999;216:72–88. doi: 10.1002/(SICI)1097-0177(199909)216:1<72::AID-DVDY9>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
- 138.Liu Z, Xu J, Colvin JS, Ornitz DM. Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes Dev. 2002;16:859–69. doi: 10.1101/gad.965602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ohbayashi N, Shibayama M, Kurotaki Y, Imanishi M, Fujimori T, Itoh N, Takada S. FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis. Genes Dev. 2002;16:870–9. doi: 10.1101/gad.965702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hurley MM, Tetradis S, Huang YF, Hock J, Kream BE, Raisz LG, Sabbieti MG. Parathyroid hormone regulates the expression of fibroblast growth factor-2 mRNA and fibroblast growth factor receptor mRNA in osteoblastic cells. J Bone Miner Res. 1999;14:776–83. doi: 10.1359/jbmr.1999.14.5.776. [DOI] [PubMed] [Google Scholar]
- 141.Sabbieti MG, Marchetti L, Abreu C, Montero A, Hand AR, Raisz LG, Hurley MM. Prostaglandins regulate the expression of fibroblast growth factor-2 in bone. Endocrinology. 1999;140:434–44. doi: 10.1210/endo.140.1.6442. [DOI] [PubMed] [Google Scholar]
- 142.Montero A, Okada Y, Tomita M, Ito M, Tsurukami H, Nakamura T, Doetschman T, Coffin JD, Hurley MM. Disruption of the fibroblast growth factor-2 gene results in decreased bone mass and bone formation. J Clin Invest. 2000;105:1085–93. doi: 10.1172/JCI8641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Xiao G, Jiang D, Gopalakrishnan R, Franceschi RT. Fibroblast growth factor 2 induction of the osteocalcin gene requires MAPK activity and phosphorylation of the osteoblast transcription factor, Cbfa1/Runx2. J Biol Chem. 2002;277:36181–7. doi: 10.1074/jbc.M206057200. [DOI] [PubMed] [Google Scholar]
- 144.Xu X, Weinstein M, Li C, Naski M, Cohen RI, Ornitz DM, Leder P, Deng C. Fibroblast growth factor receptor 2 (FGFR2)-mediated reciprocal regulation loop between FGF8 and FGF10 is essential for limb induction. Development. 1998;125:753–65. doi: 10.1242/dev.125.4.753. [DOI] [PubMed] [Google Scholar]
- 145.Eswarakumar VP, Monsonego-Ornan E, Pines M, Antonopoulou I, Morriss-Kay GM, Lonai P. The IIIc alternative of Fgfr2 is a positive regulator of bone formation. Development. 2002;129:3783–93. doi: 10.1242/dev.129.16.3783. [DOI] [PubMed] [Google Scholar]
- 146.Yu K, Xu J, Liu Z, Sosic D, Shao J, Olson EN, Towler DA, Ornitz DM. Conditional inactivation of FGF receptor 2 reveals an essential role for FGF signaling in the regulation of osteoblast function and bone growth. Development. 2003;130:3063–74. doi: 10.1242/dev.00491. [DOI] [PubMed] [Google Scholar]
- 147.Miki T, Bottaro DP, Fleming TP, Smith CL, Burgess WH, Chan AM, Aaronson SA. Determination of ligand-binding specificity by alternative splicing: two distinct growth factor receptors encoded by a single gene. Proc Natl Acad Sci U S A. 1992;89:246–50. doi: 10.1073/pnas.89.1.246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Naski MC, Ornitz DM. FGF signaling in skeletal development. Front Biosci. 1998;3:d781–94. doi: 10.2741/a321. [DOI] [PubMed] [Google Scholar]
- 149.Orr-Urtreger A, Bedford MT, Burakova T, Arman E, Zimmer Y, Yayon A, Givol D, Lonai P. Developmental localization of the splicing alternatives of fibroblast growth factor receptor-2 (FGFR2) Dev Biol. 1993;158:475–86. doi: 10.1006/dbio.1993.1205. [DOI] [PubMed] [Google Scholar]
- 150.Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol. 2001;2 doi: 10.1186/gb-2001-2-3-reviews3005. REVIEWS3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ornitz DM, Xu J, Colvin JS, McEwen DG, MacArthur CA, Coulier F, Gao G, Goldfarb M. Receptor specificity of the fibroblast growth factor family. J Biol Chem. 1996;271:15292–7. doi: 10.1074/jbc.271.25.15292. [DOI] [PubMed] [Google Scholar]
- 152.Kozawa O, Tokuda H, Matsuno H, Uematsu T. Involvement of p38 mitogen-activated protein kinase in basic fibroblast growth factor-induced interleukin-6 synthesis in osteoblasts. J Cell Biochem. 1999;74:479–85. [PubMed] [Google Scholar]
- 153.Lemonnier J, Delannoy P, Hott M, Lomri A, Modrowski D, Marie PJ. The Ser252Trp fibroblast growth factor receptor-2 (FGFR-2) mutation induces PKC-independent downregulation of FGFR-2 associated with premature calvaria osteoblast differentiation. Exp Cell Res. 2000;256:158–67. doi: 10.1006/excr.2000.4820. [DOI] [PubMed] [Google Scholar]
- 154.Lomri A, Lemonnier J, Delannoy P, Marie PJ. Increased expression of protein kinase Calpha, interleukin-1alpha, and RhoA guanosine 5'-triphosphatase in osteoblasts expressing the Ser252Trp fibroblast growth factor 2 receptor Apert mutation: identification by analysis of complementary DNA microarray. J Bone Miner Res. 2001;16:705–12. doi: 10.1359/jbmr.2001.16.4.705. [DOI] [PubMed] [Google Scholar]
- 155.Kratchmarova I, Blagoev B, Haack-Sorensen M, Kassem M, Mann M. Mechanism of divergent growth factor effects in mesenchymal stem cell differentiation. Science. 2005;308:1472–7. doi: 10.1126/science.1107627. [DOI] [PubMed] [Google Scholar]
- 156.Mundy GR, Elefteriou F. Boning up on ephrin signaling. Cell. 2006;126:441–3. doi: 10.1016/j.cell.2006.07.015. [DOI] [PubMed] [Google Scholar]
- 157.Compagni A, Logan M, Klein R, Adams RH. Control of skeletal patterning by ephrinB1-EphB interactions. Dev Cell. 2003;5:217–30. doi: 10.1016/s1534-5807(03)00198-9. [DOI] [PubMed] [Google Scholar]
- 158.Wieland I, Jakubiczka S, Muschke P, Cohen M, Thiele H, Gerlach KL, Adams RH, Wieacker P. Mutations of the ephrin-B1 gene cause craniofrontonasal syndrome. Am J Hum Genet. 2004;74:1209–15. doi: 10.1086/421532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishiwaki T, Suda T, Matsuo K. Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab. 2006;4:111–21. doi: 10.1016/j.cmet.2006.05.012. [DOI] [PubMed] [Google Scholar]
- 160.McBeath R, Pirone DM, Nelson CM, Bhadriraju K, Chen CS. Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev Cell. 2004;6:483–95. doi: 10.1016/s1534-5807(04)00075-9. [DOI] [PubMed] [Google Scholar]
- 161.Togari A. Adrenergic regulation of bone metabolism: possible involvement of sympathetic innervation of osteoblastic and osteoclastic cells. Microsc Res Tech. 2002;58:77–84. doi: 10.1002/jemt.10121. [DOI] [PubMed] [Google Scholar]
- 162.Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, Armstrong D, Ducy P, Karsenty G. Leptin regulates bone formation via the sympathetic nervous system. Cell. 2002;111:305–17. doi: 10.1016/s0092-8674(02)01049-8. [DOI] [PubMed] [Google Scholar]
- 163.Hinoi E, Fujimori S, Yoneda Y. Modulation of cellular differentiation by N-methyl-D-aspartate receptors in osteoblasts. Faseb J. 2003;17:1532–4. doi: 10.1096/fj.02-0820fje. [DOI] [PubMed] [Google Scholar]
- 164.Chien KR, Karsenty G. Longevity and lineages: toward the integrative biology of degenerative diseases in heart, muscle, and bone. Cell. 2005;120:533–44. doi: 10.1016/j.cell.2005.02.006. [DOI] [PubMed] [Google Scholar]
- 165.Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G. The molecular clock mediates leptin-regulated bone formation. Cell. 2005;122:803–15. doi: 10.1016/j.cell.2005.06.028. [DOI] [PubMed] [Google Scholar]
- 166.Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI. Identification and expression cloning of a leptin receptor, OB-R. Cell. 1995;83:1263–71. doi: 10.1016/0092-8674(95)90151-5. [DOI] [PubMed] [Google Scholar]
- 167.Wang JC, Shah N, Pantoja C, Meijsing SH, Ho JD, Scanlan TS, Yamamoto KR. Novel arylpyrazole compounds selectively modulate glucocorticoid receptor regulatory activity. Genes Dev. 2006;20:689–99. doi: 10.1101/gad.1400506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Lian JB, Javed A, Zaidi SK, Lengner C, Montecino M, van Wijnen AJ, Stein JL, Stein GS. Regulatory controls for osteoblast growth and differentiation: role of Runx/Cbfa/AML factors. Crit Rev Eukaryot Gene Expr. 2004;14:1–41. [PubMed] [Google Scholar]
- 169.Wang Q, Stacy T, Miller JD, Lewis AF, Gu TL, Huang X, Bushweller JH, Bories JC, Alt FW, Ryan G, Liu PP, Wynshaw-Boris A, Binder M, Marin-Padilla M, Sharpe AH, Speck NA. The CBFbeta subunit is essential for CBFalpha2 (AML1) function in vivo. Cell. 1996;87:697–708. doi: 10.1016/s0092-8674(00)81389-6. [DOI] [PubMed] [Google Scholar]
- 170.Kania MA, Bonner AS, Duffy JB, Gergen JP. The Drosophila segmentation gene runt encodes a novel nuclear regulatory protein that is also expressed in the developing nervous system. Genes Dev. 1990;4:1701–13. doi: 10.1101/gad.4.10.1701. [DOI] [PubMed] [Google Scholar]
- 171.Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell. 1997;89:747–54. doi: 10.1016/s0092-8674(00)80257-3. [DOI] [PubMed] [Google Scholar]
- 172.Kern B, Shen J, Starbuck M, Karsenty G. Cbfa1 contributes to the osteoblast-specific expression of type I collagen genes. J Biol Chem. 2001;276:7101–7. doi: 10.1074/jbc.M006215200. [DOI] [PubMed] [Google Scholar]
- 173.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. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997;89:755–64. doi: 10.1016/s0092-8674(00)80258-5. [DOI] [PubMed] [Google Scholar]
- 174.Ogawa E, Maruyama M, Kagoshima H, Inuzuka M, Lu J, Satake M, Shigesada K, Ito Y. PEBP2/PEA2 represents a family of transcription factors homologous to the products of the Drosophila runt gene and the human AML1 gene. Proc Natl Acad Sci U S A. 1993;90:6859–63. doi: 10.1073/pnas.90.14.6859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Banerjee C, McCabe LR, Choi JY, Hiebert SW, Stein JL, Stein GS, Lian JB. Runt homology domain proteins in osteoblast differentiation: AML3/CBFA1 is a major component of a bone-specific complex. J Cell Biochem. 1997;66:1–8. doi: 10.1002/(sici)1097-4644(19970701)66:1<1::aid-jcb1>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
- 176.Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddington RS, Mundlos S, Olsen BR, Selby PB, Owen MJ. Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell. 1997;89:765–71. doi: 10.1016/s0092-8674(00)80259-7. [DOI] [PubMed] [Google Scholar]
- 177.Mundlos S, Otto F, Mundlos C, Mulliken JB, Aylsworth AS, Albright S, Lindhout D, Cole WG, Henn W, Knoll JH, Owen MJ, Mertelsmann R, Zabel BU, Olsen BR. Mutations involving the transcription factor CBFA1 cause cleidocranial dysplasia. Cell. 1997;89:773–9. doi: 10.1016/s0092-8674(00)80260-3. [DOI] [PubMed] [Google Scholar]
- 178.Takeda S, Bonnamy JP, Owen MJ, Ducy P, Karsenty G. Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes Dev. 2001;15:467–81. doi: 10.1101/gad.845101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.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. Skeletal malformations caused by overexpression of Cbfa1 or its dominant negative form in chondrocytes. J Cell Biol. 2001;153:87–100. doi: 10.1083/jcb.153.1.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Pratap J, Galindo M, Zaidi SK, Vradii D, Bhat BM, Robinson JA, Choi JY, Komori T, Stein JL, Lian JB, Stein GS, van Wijnen AJ. Cell growth regulatory role of Runx2 during proliferative expansion of preosteoblasts. Cancer Res. 2003;63:5357–62. [PubMed] [Google Scholar]
- 181.Ducy P, Starbuck M, Priemel M, Shen J, Pinero G, Geoffroy V, Amling M, Karsenty G. A Cbfa1-dependent genetic pathway controls bone formation beyond embryonic development. Genes Dev. 1999;13:1025–36. doi: 10.1101/gad.13.8.1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Liu W, Toyosawa S, Furuichi T, Kanatani N, Yoshida C, Liu Y, Himeno M, Narai S, Yamaguchi A, Komori T. Overexpression of Cbfa1 in osteoblasts inhibits osteoblast maturation and causes osteopenia with multiple fractures. J Cell Biol. 2001;155:157–66. doi: 10.1083/jcb.200105052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Ma L, Golden S, Wu L, Maxson R. The molecular basis of Boston-type craniosynostosis: the Pro148-->His mutation in the N-terminal arm of the MSX2 homeodomain stabilizes DNA binding without altering nucleotide sequence preferences. Hum Mol Genet. 1996;5:1915–20. doi: 10.1093/hmg/5.12.1915. [DOI] [PubMed] [Google Scholar]
- 184.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. Msx2 deficiency in mice causes pleiotropic defects in bone growth and ectodermal organ formation. Nat Genet. 2000;24:391–5. doi: 10.1038/74231. [DOI] [PubMed] [Google Scholar]
- 185.Cheng SL, Shao JS, Charlton-Kachigian N, Loewy AP, Towler DA. MSX2 promotes osteogenesis and suppresses adipogenic differentiation of multipotent mesenchymal progenitors. J Biol Chem. 2003;278:45969–77. doi: 10.1074/jbc.M306972200. [DOI] [PubMed] [Google Scholar]
- 186.Ichida F, Nishimura R, Hata K, Matsubara T, Ikeda F, Hisada K, Yatani H, Cao X, Komori T, Yamaguchi A, Yoneda T. Reciprocal roles of MSX2 in regulation of osteoblast and adipocyte differentiation. J Biol Chem. 2004;279:34015–22. doi: 10.1074/jbc.M403621200. [DOI] [PubMed] [Google Scholar]
- 187.Tribioli C, Lufkin T. The murine Bapx1 homeobox gene plays a critical role in embryonic development of the axial skeleton and spleen. Development. 1999;126:5699–711. doi: 10.1242/dev.126.24.5699. [DOI] [PubMed] [Google Scholar]
- 188.Kanzler B, Kuschert SJ, Liu YH, Mallo M. Hoxa-2 restricts the chondrogenic domain and inhibits bone formation during development of the branchial area. Development. 1998;125:2587–97. doi: 10.1242/dev.125.14.2587. [DOI] [PubMed] [Google Scholar]
- 189.Chen ZF, Behringer RR. twist is required in head mesenchyme for cranial neural tube morphogenesis. Genes Dev. 1995;9:686–99. doi: 10.1101/gad.9.6.686. [DOI] [PubMed] [Google Scholar]
- 190.Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz DM, Olson EN, Justice MJ, Karsenty G. A twist code determines the onset of osteoblast differentiation. Dev Cell. 2004;6:423–35. doi: 10.1016/s1534-5807(04)00058-9. [DOI] [PubMed] [Google Scholar]
- 191.Kronenberg HM. Twist genes regulate Runx2 and bone formation. Dev Cell. 2004;6:317–8. doi: 10.1016/s1534-5807(04)00069-3. [DOI] [PubMed] [Google Scholar]
- 192.Lengner CJ, Steinman HA, Gagnon J, Smith TW, Henderson JE, Kream BE, Stein GS, Lian JB, Jones SN. Osteoblast differentiation and skeletal development are regulated by Mdm2-p53 signaling. J Cell Biol. 2006;172:909–21. doi: 10.1083/jcb.200508130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Wang X, Kua HY, Hu Y, Guo K, Zeng Q, Wu Q, Ng HH, Karsenty G, de Crombrugghe B, Yeh J, Li B. p53 functions as a negative regulator of osteoblastogenesis, osteoblast-dependent osteoclastogenesis, and bone remodeling. J Cell Biol. 2006;172:115–25. doi: 10.1083/jcb.200507106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Almog N, Rotter V. Involvement of p53 in cell differentiation and development. Biochim Biophys Acta. 1997;1333:F1–27. doi: 10.1016/s0304-419x(97)00012-7. [DOI] [PubMed] [Google Scholar]
- 195.Wu LC, Mak CH, Dear N, Boehm T, Foroni L, Rabbitts TH. Molecular cloning of a zinc finger protein which binds to the heptamer of the signal sequence for V (D)J recombination. Nucleic Acids Res. 1993;21:5067–73. doi: 10.1093/nar/21.22.5067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Oukka M, Kim ST, Lugo G, Sun J, Wu LC, Glimcher LH. A mammalian homolog of Drosophila schnurri, KRC, regulates TNF receptor-driven responses and interacts with TRAF2. Mol Cell. 2002;9:121–31. doi: 10.1016/s1097-2765(01)00434-8. [DOI] [PubMed] [Google Scholar]
- 197.Jones DC, Wein MN, Oukka M, Hofstaetter JG, Glimcher MJ, Glimcher LH. Regulation of adult bone mass by the zinc finger adapter protein Schnurri-3. Science. 2006;312:1223–7. doi: 10.1126/science.1126313. [DOI] [PubMed] [Google Scholar]
- 198.Yoshida CA, Furuichi T, Fujita T, Fukuyama R, Kanatani N, Kobayashi S, Satake M, Takada K, Komori T. Core-binding factor beta interacts with Runx2 and is required for skeletal development. Nat Genet. 2002;32:633–8. doi: 10.1038/ng1015. [DOI] [PubMed] [Google Scholar]
- 199.Kundu M, Javed A, Jeon JP, Horner A, Shum L, Eckhaus M, Muenke M, Lian JB, Yang Y, Nuckolls GH, Stein GS, Liu PP. Cbfbeta interacts with Runx2 and has a critical role in bone development. Nat Genet. 2002;32:639–44. doi: 10.1038/ng1050. [DOI] [PubMed] [Google Scholar]
- 200.Miller J, Horner A, Stacy T, Lowrey C, Lian JB, Stein G, Nuckolls GH, Speck NA. The core-binding factor beta subunit is required for bone formation and hematopoietic maturation. Nat Genet. 2002;32:645–9. doi: 10.1038/ng1049. [DOI] [PubMed] [Google Scholar]
- 201.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. 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. 2003;23:3339–51. doi: 10.1128/MCB.23.9.3339-3351.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Miyazono K, ten Dijke P, Heldin CH. TGF-beta signaling by Smad proteins. Adv Immunol. 2000;75:115–57. doi: 10.1016/s0065-2776(00)75003-6. [DOI] [PubMed] [Google Scholar]
- 203.Pelletier N, Champagne N, Stifani S, Yang XJ. MOZ and MORF histone acetyltransferases interact with the Runt-domain transcription factor Runx2. Oncogene. 2002;21:2729–40. doi: 10.1038/sj.onc.1205367. [DOI] [PubMed] [Google Scholar]
- 204.Thomas T, Voss AK, Chowdhury K, Gruss P. Querkopf, a MYST family histone acetyltransferase, is required for normal cerebral cortex development. Development. 2000;127:2537–48. doi: 10.1242/dev.127.12.2537. [DOI] [PubMed] [Google Scholar]
- 205.Thomas DM, Carty SA, Piscopo DM, Lee JS, Wang WF, Forrester WC, Hinds PW. The retinoblastoma protein acts as a transcriptional coactivator required for osteogenic differentiation. Mol Cell. 2001;8:303–16. doi: 10.1016/s1097-2765(01)00327-6. [DOI] [PubMed] [Google Scholar]
- 206.Cui CB, Cooper LF, Yang X, Karsenty G, Aukhil I. Transcriptional coactivation of bone-specific transcription factor Cbfa1 by TAZ. Mol Cell Biol. 2003;23:1004–13. doi: 10.1128/MCB.23.3.1004-1013.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Wang W, Wang YG, Reginato AM, Glotzer DJ, Fukai N, Plotkina S, Karsenty G, Olsen BR. Groucho homologue Grg5 interacts with the transcription factor Runx2-Cbfa1 and modulates its activity during postnatal growth in mice. Dev Biol. 2004;270:364–81. doi: 10.1016/j.ydbio.2004.03.003. [DOI] [PubMed] [Google Scholar]
- 208.Hong JH, Hwang ES, McManus MT, Amsterdam A, Tian Y, Kalmukova R, Mueller E, Benjamin T, Spiegelman BM, Sharp PA, Hopkins N, Yaffe MB. TAZ, a transcriptional modulator of mesenchymal stem cell differentiation. Science. 2005;309:1074–8. doi: 10.1126/science.1110955. [DOI] [PubMed] [Google Scholar]
- 209.Peterson CL, Laniel MA. Histones and histone modifications. Curr Biol. 2004;14:R546–51. doi: 10.1016/j.cub.2004.07.007. [DOI] [PubMed] [Google Scholar]
- 210.Westendorf JJ, Zaidi SK, Cascino JE, Kahler R, van Wijnen AJ, Lian JB, Yoshida M, Stein GS, Li X. Runx2 (Cbfa1, AML-3) interacts with histone deacetylase 6 and represses the p21 (CIP1/WAF1) promoter. Mol Cell Biol. 2002;22:7982–92. doi: 10.1128/MCB.22.22.7982-7992.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Schroeder TM, Kahler RA, Li X, Westendorf JJ. Histone deacetylase 3 interacts with runx2 to repress the osteocalcin promoter and regulate osteoblast differentiation. J Biol Chem. 2004;279:41998–2007. doi: 10.1074/jbc.M403702200. [DOI] [PubMed] [Google Scholar]
- 212.Vega RB, Matsuda K, Oh J, Barbosa AC, Yang X, Meadows E, McAnally J, Pomajzl C, Shelton JM, Richardson JA, Karsenty G, Olson EN. Histone deacetylase 4 controls chondrocyte hypertrophy during skeletogenesis. Cell. 2004;119:555–66. doi: 10.1016/j.cell.2004.10.024. [DOI] [PubMed] [Google Scholar]
- 213.Schroeder TM, Westendorf JJ. Histone deacetylase inhibitors promote osteoblast maturation. J Bone Miner Res. 2005;20:2254–63. doi: 10.1359/JBMR.050813. [DOI] [PubMed] [Google Scholar]
- 214.Cho HH, Park HT, Kim YJ, Bae YC, Suh KT, Jung JS. Induction of osteogenic differentiation of human mesenchymal stem cells by histone deacetylase inhibitors. J Cell Biochem. 2005;96:533–42. doi: 10.1002/jcb.20544. [DOI] [PubMed] [Google Scholar]
- 215.Jeon EJ, Lee KY, Choi NS, Lee MH, Kim HN, Jin YH, Ryoo HM, Choi JY, Yoshida M, Nishino N, Oh BC, Lee KS, Lee YH, Bae SC. Bone morphogenetic protein-2 stimulates Runx2 acetylation. J Biol Chem. 2006;281:16502–11. doi: 10.1074/jbc.M512494200. [DOI] [PubMed] [Google Scholar]
- 216.Thirunavukkarasu K, Mahajan M, McLarren KW, Stifani S, Karsenty G. Two domains unique to osteoblast-specific transcription factor Osf2/Cbfa1 contribute to its transactivation function and its inability to heterodimerize with Cbfbeta. Mol Cell Biol. 1998;18:4197–208. doi: 10.1128/mcb.18.7.4197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Javed A, Guo B, Hiebert S, Choi JY, Green J, Zhao SC, Osborne MA, Stifani S, Stein JL, Lian JB, van Wijnen AJ, Stein GS. Groucho/TLE/R-esp proteins associate with the nuclear matrix and repress RUNX (CBF (alpha)/AML/PEBP2 (alpha)) dependent activation of tissue-specific gene transcription. J Cell Sci. 2000;113(Pt 12):2221–31. doi: 10.1242/jcs.113.12.2221. [DOI] [PubMed] [Google Scholar]
- 218.Zaidi SK, Sullivan AJ, Medina R, Ito Y, van Wijnen AJ, Stein JL, Lian JB, Stein GS. Tyrosine phosphorylation controls Runx2-mediated subnuclear targeting of YAP to repress transcription. Embo J. 2004;23:790–9. doi: 10.1038/sj.emboj.7600073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Tintut Y, Parhami F, Le V, Karsenty G, Demer LL. Inhibition of osteoblast-specific transcription factor Cbfa1 by the cAMP pathway in osteoblastic cells. Ubiquitin/proteasome-dependent regulation. J Biol Chem. 1999;274:28875–9. doi: 10.1074/jbc.274.41.28875. [DOI] [PubMed] [Google Scholar]
- 220.Zhao M, Qiao M, Oyajobi BO, Mundy GR, Chen D. E3 ubiquitin ligase Smurf1 mediates core-binding factor alpha1/Runx2 degradation and plays a specific role in osteoblast differentiation. J Biol Chem. 2003;278:27939–44. doi: 10.1074/jbc.M304132200. [DOI] [PubMed] [Google Scholar]
- 221.Shen R, Chen M, Wang YJ, Kaneki H, Xing L, O'Keefe R J, Chen D. Smad6 interacts with Runx2 and mediates Smad ubiquitin regulatory factor 1-induced Runx2 degradation. J Biol Chem. 2006;281:3569–76. doi: 10.1074/jbc.M506761200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Kaneki H, Guo R, Chen D, Yao Z, Schwarz EM, Zhang YE, Boyce BF, Xing L. Tumor necrosis factor promotes Runx2 degradation through up-regulation of Smurf1 and Smurf2 in osteoblasts. J Biol Chem. 2006;281:4326–33. doi: 10.1074/jbc.M509430200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Zhao M, Qiao M, Harris SE, Oyajobi BO, Mundy GR, Chen D. Smurf1 inhibits osteoblast differentiation and bone formation in vitro and in vivo. J Biol Chem. 2004;279:12854–9. doi: 10.1074/jbc.M313294200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.D'Alonzo RC, Selvamurugan N, Karsenty G, Partridge NC. Physical interaction of the activator protein-1 factors c-Fos and c-Jun with Cbfa1 for collagenase-3 promoter activation. J Biol Chem. 2002;277:816–22. doi: 10.1074/jbc.M107082200. [DOI] [PubMed] [Google Scholar]
- 225.Hanai J, Chen LF, Kanno T, Ohtani-Fujita N, Kim WY, Guo WH, Imamura T, Ishidou Y, Fukuchi M, Shi MJ, Stavnezer J, Kawabata M, Miyazono K, Ito Y. Interaction and functional cooperation of PEBP2/CBF with Smads. Synergistic induction of the immunoglobulin germline Calpha promoter. J Biol Chem. 1999;274:31577–82. doi: 10.1074/jbc.274.44.31577. [DOI] [PubMed] [Google Scholar]
- 226.Nishimura R, Hata K, Harris SE, Ikeda F, Yoneda T. Core-binding factor alpha 1 (Cbfa1) induces osteoblastic differentiation of C2C12 cells without interactions with Smad1 and Smad5. Bone. 2002;31:303–12. doi: 10.1016/s8756-3282(02)00826-8. [DOI] [PubMed] [Google Scholar]
- 227.Sato M, Morii E, Komori T, Kawahata H, Sugimoto M, Terai K, Shimizu H, Yasui T, Ogihara H, Yasui N, Ochi T, Kitamura Y, Ito Y, Nomura S. Transcriptional regulation of osteopontin gene in vivo by PEBP2alphaA/CBFA1 and ETS1 in the skeletal tissues. Oncogene. 1998;17:1517–25. doi: 10.1038/sj.onc.1202064. [DOI] [PubMed] [Google Scholar]
- 228.Gutierrez S, Javed A, Tennant DK, van Rees M, Montecino M, Stein GS, Stein JL, Lian JB. CCAAT/enhancer-binding proteins (C/EBP) beta and delta activate osteocalcin gene transcription and synergize with Runx2 at the C/EBP element to regulate bone-specific expression. J Biol Chem. 2002;277:1316–23. doi: 10.1074/jbc.M106611200. [DOI] [PubMed] [Google Scholar]
- 229.Hata K, Nishimura R, Ueda M, Ikeda F, Matsubara T, Ichida F, Hisada K, Nokubi T, Yamaguchi A, Yoneda T. A CCAAT/enhancer binding protein beta isoform, liver-enriched inhibitory protein, regulates commitment of osteoblasts and adipocytes. Mol Cell Biol. 2005;25:1971–9. doi: 10.1128/MCB.25.5.1971-1979.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Shirakabe K, Terasawa K, Miyama K, Shibuya H, Nishida E. Regulation of the activity of the transcription factor Runx2 by two homeobox proteins, Msx2 and Dlx5. Genes Cells. 2001;6:851–6. doi: 10.1046/j.1365-2443.2001.00466.x. [DOI] [PubMed] [Google Scholar]
- 231.Hassan MQ, Javed A, Morasso MI, Karlin J, Montecino M, van Wijnen AJ, Stein GS, Stein JL, Lian JB. Dlx3 transcriptional regulation of osteoblast differentiation: temporal recruitment of Msx2, Dlx3, and Dlx5 homeodomain proteins to chromatin of the osteocalcin gene. Mol Cell Biol. 2004;24:9248–61. doi: 10.1128/MCB.24.20.9248-9261.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.McLarren KW, Lo R, Grbavec D, Thirunavukkarasu K, Karsenty G, Stifani S. The mammalian basic helix loop helix protein HES-1 binds to and modulates the transactivating function of the runt-related factor Cbfa1. J Biol Chem. 2000;275:530–8. doi: 10.1074/jbc.275.1.530. [DOI] [PubMed] [Google Scholar]
- 233.Sowa H, Kaji H, Hendy GN, Canaff L, Komori T, Sugimoto T, Chihara K. Menin is required for bone morphogenetic protein 2- and transforming growth factor beta-regulated osteoblastic differentiation through interaction with Smads and Runx2. J Biol Chem. 2004;279:40267–75. doi: 10.1074/jbc.M401312200. [DOI] [PubMed] [Google Scholar]
- 234.Newberry EP, Latifi T, Towler DA. Reciprocal regulation of osteocalcin transcription by the homeodomain proteins Msx2 and Dlx5. Biochemistry. 1998;37:16360–8. doi: 10.1021/bi981878u. [DOI] [PubMed] [Google Scholar]
- 235.Selvamurugan N, Chou WY, Pearman AT, Pulumati MR, Partridge NC. Parathyroid hormone regulates the rat collagenase-3 promoter in osteoblastic cells through the cooperative interaction of the activator protein-1 site and the runt domain binding sequence. J Biol Chem. 1998;273:10647–57. doi: 10.1074/jbc.273.17.10647. [DOI] [PubMed] [Google Scholar]
- 236.Afzal F, Pratap J, Ito K, Ito Y, Stein JL, van Wijnen AJ, Stein GS, Lian JB, Javed A. Smad function and intranuclear targeting share a Runx2 motif required for osteogenic lineage induction and BMP2 responsive transcription. J Cell Physiol. 2005;204:63–72. doi: 10.1002/jcp.20258. [DOI] [PubMed] [Google Scholar]
- 237.Kahler RA, Westendorf JJ. Lymphoid enhancer factor-1 and beta-catenin inhibit Runx2-dependent transcriptional activation of the osteocalcin promoter. J Biol Chem. 2003;278:11937–44. doi: 10.1074/jbc.M211443200. [DOI] [PubMed] [Google Scholar]
- 238.Jeon MJ, Kim JA, Kwon SH, Kim SW, Park KS, Park SW, Kim SY, Shin CS. Activation of peroxisome proliferator-activated receptor-gamma inhibits the Runx2-mediated transcription of osteocalcin in osteoblasts. J Biol Chem. 2003;278:23270–7. doi: 10.1074/jbc.M211610200. [DOI] [PubMed] [Google Scholar]
- 239.Alliston T, Choy L, Ducy P, Karsenty G, Derynck R. TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. Embo J. 2001;20:2254–72. doi: 10.1093/emboj/20.9.2254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Zamurovic N, Cappellen D, Rohner D, Susa M. Coordinated activation of notch, Wnt, and transforming growth factor-beta signaling pathways in bone morphogenic protein 2-induced osteogenesis. Notch target gene Hey1 inhibits mineralization and Runx2 transcriptional activity. J Biol Chem. 2004;279:37704–15. doi: 10.1074/jbc.M403813200. [DOI] [PubMed] [Google Scholar]
- 241.Kim S, Koga T, Isobe M, Kern BE, Yokochi T, Chin YE, Karsenty G, Taniguchi T, Takayanagi H. Stat1 functions as a cytoplasmic attenuator of Runx2 in the transcriptional program of osteoblast differentiation. Genes Dev. 2003;17:1979–91. doi: 10.1101/gad.1119303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.McCarthy TL, Ji C, Chen Y, Kim KK, Imagawa M, Ito Y, Centrella M. Runt domain factor (Runx)-dependent effects on CCAAT/enhancer-binding protein delta expression and activity in osteoblasts. J Biol Chem. 2000;275:21746–53. doi: 10.1074/jbc.M002291200. [DOI] [PubMed] [Google Scholar]
- 243.Milona MA, Gough JE, Edgar AJ. Expression of alternatively spliced isoforms of human Sp7 in osteoblast-like cells. BMC Genomics. 2003;4(43) doi: 10.1186/1471-2164-4-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Celil AB, Campbell PG. BMP-2 and insulin-like growth factor-I mediate Osterix (Osx) expression in human mesenchymal stem cells via the MAPK and protein kinase D signaling pathways. J Biol Chem. 2005;280:31353–9. doi: 10.1074/jbc.M503845200. [DOI] [PubMed] [Google Scholar]
- 245.Koga T, Matsui Y, Asagiri M, Kodama T, de Crombrugghe B, Nakashima K, Takayanagi H. NFAT and Osterix cooperatively regulate bone formation. Nat Med. 2005;11:880–5. doi: 10.1038/nm1270. [DOI] [PubMed] [Google Scholar]
- 246.Deckelbaum RA, Majithia A, Booker T, Henderson JE, Loomis CA. The homeoprotein engrailed 1 has pleiotropic functions in calvarial intramembranous bone formation and remodeling. Development. 2006;133:63–74. doi: 10.1242/dev.02171. [DOI] [PubMed] [Google Scholar]
- 247.Fedde KN, Blair L, Silverstein J, Coburn SP, Ryan LM, Weinstein RS, Waymire K, Narisawa S, Millan JL, MacGregor GR, Whyte MP. Alkaline phosphatase knock-out mice recapitulate the metabolic and skeletal defects of infantile hypophosphatasia. J Bone Miner Res. 1999;14:2015–26. doi: 10.1359/jbmr.1999.14.12.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.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. ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry Syndrome. Cell. 2004;117:387–98. doi: 10.1016/s0092-8674(04)00344-7. [DOI] [PubMed] [Google Scholar]
- 249.Yang X, Karsenty G. ATF4, the osteoblast accumulation of which is determined post-translationally, can induce osteoblast-specific gene expression in nonosteoblastic cells. J Biol Chem. 2004;279:47109–14. doi: 10.1074/jbc.M410010200. [DOI] [PubMed] [Google Scholar]
- 250.Xiao G, Jiang D, Ge C, Zhao Z, Lai Y, Boules H, Phimphilai M, Yang X, Karsenty G, Franceschi RT. Cooperative interactions between activating transcription factor 4 and Runx2/Cbfa1 stimulate osteoblast-specific osteocalcin gene expression. J Biol Chem. 2005;280:30689–96. doi: 10.1074/jbc.M500750200. [DOI] [PubMed] [Google Scholar]
- 251.Trainor PA, Krumlauf R. Hox genes, neural crest cells and branchial arch patterning. Curr Opin Cell Biol. 2001;13:698–705. doi: 10.1016/s0955-0674(00)00273-8. [DOI] [PubMed] [Google Scholar]
- 252.Ellies DL, Krumlauf R. Bone formation: The nuclear matrix reloaded. Cell. 2006;125:840–2. doi: 10.1016/j.cell.2006.05.022. [DOI] [PubMed] [Google Scholar]
- 253.Dobreva G, Chahrour M, Dautzenberg M, Chirivella L, Kanzler B, Farinas I, Karsenty G, Grosschedl R. SATB2 is a multifunctional determinant of craniofacial patterning and osteoblast differentiation. Cell. 2006;125:971–86. doi: 10.1016/j.cell.2006.05.012. [DOI] [PubMed] [Google Scholar]
- 254.Dobreva G, Dambacher J, Grosschedl R. SUMO modification of a novel MAR-binding protein, SATB2, modulates immunoglobulin mu gene expression. Genes Dev. 2003;17:3048–61. doi: 10.1101/gad.1153003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Britanova O, Akopov S, Lukyanov S, Gruss P, Tarabykin V. Novel transcription factor Satb2 interacts with matrix attachment region DNA elements in a tissue-specific manner and demonstrates cell-type-dependent expression in the developing mouse CNS. Eur J Neurosci. 2005;21:658–68. doi: 10.1111/j.1460-9568.2005.03897.x. [DOI] [PubMed] [Google Scholar]
- 256.FitzPatrick DR, Carr IM, McLaren L, Leek JP, Wightman P, Williamson K, Gautier P, McGill N, Hayward C, Firth H, Markham AF, Fantes JA, Bonthron DT. Identification of SATB2 as the cleft palate gene on 2q32-q33. Hum Mol Genet. 2003;12:2491–501. doi: 10.1093/hmg/ddg248. [DOI] [PubMed] [Google Scholar]
- 257.Behrens A, Haigh J, Mechta-Grigoriou F, Nagy A, Yaniv M, Wagner EF. Impaired intervertebral disc formation in the absence of Jun. Development. 2003;130:103–9. doi: 10.1242/dev.00186. [DOI] [PubMed] [Google Scholar]
- 258.Kenner L, Hoebertz A, Beil T, Keon N, Karreth F, Eferl R, Scheuch H, Szremska A, Amling M, Schorpp-Kistner M, Angel P, Wagner EF. Mice lacking JunB are osteopenic due to cell-autonomous osteoblast and osteoclast defects. J Cell Biol. 2004;164:613–23. doi: 10.1083/jcb.200308155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Grigoriadis AE, Schellander K, Wang ZQ, Wagner EF. Osteoblasts are target cells for transformation in c-fos transgenic mice. J Cell Biol. 1993;122:685–701. doi: 10.1083/jcb.122.3.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Eferl R, Hoebertz A, Schilling AF, Rath M, Karreth F, Kenner L, Amling M, Wagner EF. The Fos-related antigen Fra-1 is an activator of bone matrix formation. Embo J. 2004;23:2789–99. doi: 10.1038/sj.emboj.7600282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Robledo RF, Rajan L, Li X, Lufkin T. The Dlx5 and Dlx6 homeobox genes are essential for craniofacial, axial, and appendicular skeletal development. Genes Dev. 2002;16:1089–101. doi: 10.1101/gad.988402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Galceran J, Sustmann C, Hsu SC, Folberth S, Grosschedl R. LEF1-mediated regulation of Delta-like1 links Wnt and Notch signaling in somitogenesis. Genes Dev. 2004;18:2718–23. doi: 10.1101/gad.1249504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Lecka-Czernik B, Gubrij I, Moerman EJ, Kajkenova O, Lipschitz DA, Manolagas SC, Jilka RL. Inhibition of Osf2/Cbfa1 expression and terminal osteoblast differentiation by PPARgamma2. J Cell Biochem. 1999;74:357–71. [PubMed] [Google Scholar]
- 264.Brault V, Moore R, Kutsch S, Ishibashi M, Rowitch DH, McMahon AP, Sommer L, Boussadia O, Kemler R. Inactivation of the beta-catenin gene by Wnt1-Cre-mediated deletion results in dramatic brain malformation and failure of craniofacial development. Development. 2001;128:1253–64. doi: 10.1242/dev.128.8.1253. [DOI] [PubMed] [Google Scholar]