Você está na página 1de 25

Periodontology 2000, Vol. 41, 2006, 4872 Printed in Singapore.

All rights reserved

Copyright Blackwell Munksgaard 2006

PERIODONTOLOGY 2000

Effects of growth factors and cytokines on osteoblast differentiation


F R A N C I S J. H U G H E S , W E N D Y T U R N E R , G E O R G I O S B E L I B A S A K I S & GIANLUCA MARTUSCELLI

The therapeutic management of new bone formation remains one of the key issues in periodontology and dental implantology. A full understanding of the nature of the bone-forming cells (osteoblasts) and their lineage, the factors that may regulate osteoblast behavior and bone formation locally, and how these different regulatory mechanisms could interact, has been the subject of extensive study for many years, particularly with the aspiration that such an understanding will lead to the development of novel biologically based therapies for the management of bone regeneration, not only in dental applications but also for many other disciplines, such as orthopedic surgery, and maxillofacial and craniofacial surgery. The aim of this review was as follows. to describe the nature of the osteoblast phenotype, its lineage and the genetic regulation of osteoblast differentiation and function. to discuss the role of a range of cytokines and growth factors in regulating osteoblast differentiation and function. to review how some of these cytokine pathways may be regulated and interact with each other in the co-ordinated control of osteoblast function. to review, briey, studies that have investigated the pharmacological application of cytokines for therapeutic management of bone formation locally in vivo. Cytokines are soluble, secreted glycoproteins which act as local signaling molecules to control and co-ordinate cellular behavior and function. They function as ligands which bind to cell-surface receptors, triggering a series of intracellular signaling events, ultimately resulting in the modulation of gene expression in their target cells. Growth and differentiation factors are generally considered to be a subset

of cytokines with anabolic physiological and pathophysiological roles in the regulation of tissue growth and healing. Although this terminology is widely used, the distinction between growth factors and cytokines is probably an articial one, particularly given the different complex roles that these molecules may all have on cell and tissue function. However in keeping with convention, here we have used the term growth factors to describe this distinct subgroup of cytokines, and have separately considered the role of cytokines which are particularly associated with inammation on bone cell regulation.

Osteoblast lineage and function


The osteoblast phenotype
Osteoblasts are specialized mesenchymal-derived cells whose function is the deposition and maintenance of skeletal tissues. As with any cell type, they are dened ultimately by their expression of genes that are a subset of the total genome and which dene their unique function. Specically, osteoblasts express the genes coding for the bone matrix proteins, high levels of the tissue nonspecic form of alkaline phosphatase and are also frequently characterized by their expression of parathyroid hormone/parathyroid hormone receptors, which results in a typical cAMP response on stimulation with the osteotropic parathyroid hormone. The bone matrix proteins include type I collagen and a number of other matrix proteins, which to a greater or lesser extent are characteristic or specically expressed in bone. Collagen type I makes up 90% of

48

Effects of growth factors and cytokines on osteoblast differentiation

the organic matrix of bone and provides the strength, structure and elasticity of the mature bone tissue. The noncollagenous proteins of bone include osteocalcin (bone GLA protein) osteopontin, osteonectin and bone sialoprotein. In addition, the proteoglycans decorin and biglycan are signicant components of the bone organic matrix. Although a full description of the function of the different matrix constituents of bone is beyond the scope of this review, these proteins collectively have calcium-binding activity, which is probably responsible for the exquisite regulation of hydroxyapatite crystal deposition, orientation and specic crystal size seen in the mineralized bone matrix. In addition, it is known that bone sialoprotein and osteopontin contain the ArgGlyAsp (RGD) peptide cell-attachment sequences and can mediate attachment of both osteoblasts and osteoclasts to the bone matrix. Of the noncollagenous proteins of bone, osteocalcin is considered to be the most bone-specic. Expression of osteocalcin in the mature organism is restricted to bone, dentin and cementum. Evidence particularly from mouse knockout models suggest that the bone matrix proteins play an important role in regulating the overall control of bone mass. For example, the osteocalcin knockout mouse exhibits an overall increase in bone mass as its main phenotype (54). The expression of bone-specic genes and proteins are valuable markers for demonstration of bone osteoblast phenotype in vitro, and understanding of the transcriptional regulation of these genes is of fundamental importance in the regulation of osteoblast differentiation and function.

The osteoblast lineage


Osteoblasts are derived from multipotent mesenchymal stem cells that give rise to progenitor cells of restricted osteoblast lineage and which may then undergo a series of proliferation/amplication stages before expressing recognizable specic osteoblastic markers. Early studies of the kinetics of normal osteoblast turnover using pulse-chase 3H-thymidine labeling demonstrate the recruitment of osteoprogenitor cells from the surrounding connective tissues toward the periosteal layer on the surface of the bone (188, 189). The periosteum consists of two layers in these studies: a broperiosteum on the outside most distant from the bone surface; and an inner osteogenic periosteum where the rst directly recognizable cell of the differentiating osteoblast lineage, the preosteoblast, is found. Pre-osteoblasts retain a prolif-

erative capcacity but express many of the proteins associated with the mature osteoblast phenotype, including alkaline phosphatase and osteopontin. The mature osteoblast layer lies immediately adjacent to the bone and is a post-mitotic cell expressing all of the features of the mature phenotype (189). These descriptions give rise to the concept of recruitment of proliferating osteoprogenitor cells from adjacent connective tissues, which undergo further differentiation, resulting in the expression of the mature phenotype. These studies also suggest that mature osteoblasts have an average lifespan of 1 month, after which either they undergo apoptosis, to be replaced by newly differentiated osteoblasts or alternatively about one-third of these cells may be incorporated into deposited bone matrix as osteocytes. The osteocyte is thus the most differentiated cell of the osteoblast lineage and may persist in bone matrix for very lengthy periods of time. Although its functions are not fully understood, there is compelling evidence that the osteocyte has a critical role in the maintenance of bone mass and is the principal bone cell responsible for mechanotransduction. It is well established that maintenance of bone mass relies on regular mechanical stimulation. It is thought that the subtle exing seen in bones in response to any loading causes pulsatile uid ow through osteocyte canaliculi, resulting in signal transduction by osteocytes in order to retain bone mass (24, 128, 228). The precise molecular mechanisms whereby this occurs are unknown. However, mechanical strain and pulsatile uid ow are able to induce signaling molecules, such as nitric oxide, by osteocytes in vitro (127, 194). Interestingly, the recently described bone morphogenetic protein (BMP) inhibitor, sclerostin, has also been localized specically to osteocytes and may contribute to the regulation of overall bone mass (264). Around the periodontal tissues, similar pulse chase 3H-thymidine-labeling experiments demonstrate the continual recruitment of proliferating cells from bone marrow stroma through vascular channels that communicate with the periodontal ligament. These cells appear to preferentially migrate to the osteoblast (and cementoblast) surfaces, consistent with the recruitment of osteoprogenitor cells from stem cells located in the bone marrow stroma through to the periodontal ligament and ultimately undergo osteoblastic differentiation, akin to the kinetics described in other bony tissues (159). Thus, the periodontal ligament is constantly repopulated by relatively undifferentiated mesenchymal cells and is a

49

Hughes et al.

rich source of progenitor cells capable of giving rise to new bone formation (see Fig. 1).

Mesenchymal stem cells


Stem cells are dened as undifferentiated selfrenewing cells. In the mature organism they tend to occur with relatively low frequency and although, by denition, they have extremely high proliferative potential, they are typically not continually cycling. In addition, many (but not all) stem cells have multiple potentials, that is they have the capacity to give rise to a range of differentiated phenotypes. The existence of undifferentiated self-renewing cells is clearly essential for the organism to replenish continually differentiated cells which have reached the end of their normal lifespan. Classic studies by Owen & Fredenstein were the rst to describe the existence of multipotent mesenchymal stem cells isolated from bone marrow stromal cell populations (66, 67, 190). In these experiments it was demonstrated that colonies from bone marrow stroma (and therefore of clonal origin) could give rise to a wide range of connective tissues, including bone, cartilage, muscle, brous and adipose tissues, when re-implanted into a suitable host animal. More formal demonstration of the true multipotential nature of mesenchymal stem cells was subsequently demonstrated in clonally derived cell lines in vitro including the murine embryonic broblastic cell line, C3H10T1/2 (132), and the rat calvaria-derived line, RCJ3.1 (84). These and other multipotent mesenchymal cell lines have been valuable tools for using to investigate the role of a range of different factors that may regulate the commitment of multipotent stem cells to cells of restricted lineages, such as osteoprogenitor cells. In recent years there has been considerable debate about the plasticity of stem cells, that is, the ability of committed progenitor cells to dedifferentiate and subsequently redifferentiate along a different lineage. There is considerable evidence for the existence of bipotent osteoblastadipocyte cells from bone marrow in both human and other species (4, 18, 42) and these also exhibit extensive potential for undergoing cell renewal. These data suggest the possibility that restriction to specic differentiation pathways may not occur as a single event but it is possible for multipotent stem cells to exhibit increasing lineagerestricted specication. Further evidence of this is provided by studies of C3H10T1/2 cells, where subclones have also been shown to exhibit tripotent properties (132) and these potentials have also been

Fig. 1. Demonstration of continual recruitment of progenitor cells from bone marrow to the periodontal ligament. Cycling cells were labeled by a bromodeoxyuridine (BrdU) pulse on day 0 and followed-up for periods of up to 10 days in Wistar rats. (A) Day 0. A bone marrow space is seen within the bone, communicating directly with the PDL via a vascular channel. Labeled cycling cells (red) are largely restricted to the bone marrow stroma. (B) Day 6. Labeled cells have now migrated into the periodontal tissue spaces and are seen adjacent to vascular channel openings into the ligament. (C) Day 10. Labeled cells are relatively evenly distributed throughout the ligament, tending toward the bony surface. B, bone; T, tooth; PDL, periodontal ligament. Original magnication 100 [King GN, King N, Hughes FJ, unpublished, based on experiments described by McCulloch et al. (159)].

50

Effects of growth factors and cytokines on osteoblast differentiation

reported in primary marrow stromal cells, particularly as osteoblast/chondrocyte/adipocyte stem cells (2, 187, 195). There is much less clear evidence of cells at an advanced stage of differentiation exhibiting true dedifferentiation and redifferentiation along distinct pathways (132). However, studies suggest that this phenomenon may occur in osteoblasts redifferentiating into chondrocytes. In support of the possibility of a differentiated cell subsequently redifferentiating as a distinct phenotype, it has been shown that osteoblastic cells, when transfected with the tissue-specic transcription factor, PPAR-c 2, can be converted to express an adipocytic phenotype (4). In addition to these complexities of the stem cell system, it has proved extremely difcult to identify specic cells of the mesenchymal lineages with labels that specically reect a particular stage in the stem cell/early commitment phases of connective tissue cell lineages. Typically, in stem cell studies using culture methods, in vitro differentiation potentials are elucidated by directly demonstrating differentiation outcomes and ascribing those to cultured cells in retrospect. In addition, at least one antibody (Stro1) has proved extremely useful in identifying the cells in marrow stromal cell cultures with osteogenic potential (225). Stro-1 staining in combination with alkaline phosphatase expression using uorescenceactivated cell sorting has demonstrated the presence of early osteogenic precursor cells in a Stro-1-positive/alkaline phosphatase-negative population of marrow stromal cells (51, 85, 222). During differentiation it has been shown that Stro-1 expression is lost, whilst alkaline phosphatase expression is upregulated (257). Despite these observations and many other attempts, it has still proven difcult and impossible at the present time to dene a marker or markers that would unequivocally identify a single multipotent mesenchymal stem cell in a mixed stromal cell population.

Transcriptional regulation of osteoblastic commitment and differentiation


Lineage-specic gene expression is ultimately under the control of transcription factors that act to regulate specic gene expression. They act as the key switching mechanisms to induce gene transcription. Considerable progress has been made in identifying those transcription factors which act as master switches during commitment of multipotent cells to specic lineages. Basic helix-loop-helix genes of the myoD family are known to specify commitment and differentiation of mesenchymal stem cells to the

myoblast lineage (260). Similarly, the transcription factor, ppar-c 2, can specify adipocyte differentiation (209), and sox-9 expression is required for chondrocyte differentiation (44). A major breakthrough in the understanding of genetic regulation of osteoblast differentiation was made with the identication of the role of the transcription factor core binding factor 1 (Cbfa-1/Runx-2) (55, 130). Cbfa-1/Runx-2 expression is an absolute requirement for osteoblast differentiation. In cbfa-1 knockout mice there is a normal cartilaginous skeleton seen but a complete absence of bone (and tooth) formation. Cbfa-1/Runx2 is known to interact directly with the osteocalcin promoter to induce its expression (56). However an additional transcription factor, osterix, which is a downstream target for Cbfa-1/Runx-2, has also been shown to be an absolute requirement for normal osteoblast differentiation in knockout mice experiments (170). More recent studies have shown the existence of distinct isoforms of Cbfa-1, which may have subtly different roles during normal tissue formation, including regulation of cartilage expression in addition to bone. The expression of Cbfa-1/Runx-2 occurs quite late in the osteoblast lineage scheme and physiologically probably does not specify osteoblastic commitment events in multipotent stem cell populations. Recently, the role of the transcription factor, TAZ, has been reported, which appears to act by specifying osteoblastic cell fate in bipotent osteoblast/adipocyte stem cells, by activating Cbfa-1/Runx-2 whilst simultaneously repressing the adipocyte transcription factor, ppar-c 2 (95), Experimental overexpression of TAZ in primary mesenchymal stem cells appears to preferentially specify osteoblastic differentiation in mesenchymal stem cells. A number of other transcription factors, whilst not being absolute requirements for osteoblast differentiation, are also known to regulate osteoblast differentiation. These include Msx-2, Dlx-5 and the AP-1 family of transcription factors formed by heterodimers of members of fos and jun families. Although a full review of these molecules is beyond the scope of this review, msx-2 appears to act upstream of Cbfa-1/Runx-2 (91) whilst Dlx-5 may be a transcriptional regulator of late stage osteoblast differentiation acting downstream of Cbfa-1/Runx-2.

Summary
In summary, the osteoblast lineage consists of mesenchymal stem cells which exhibit self-renewal and may include both multipotent cells and those of

51

Hughes et al.

Fig. 2. Schematic diagram of stages in the osteoblast lineage and some phenotypic features seen at different stages of the lineage. Cbfa-1, transcription factor expression. AIP, alkaline phosphatase; AP-1, activator-protein-1; BSP, bone sialoprotein; OSX, osterix; PTH, parathyroid hormone; PTHrP R, PTH-related protein receptor.

more restricted potentials. Commitment of these cells to the osteoblast lineage is likely to be mediated by the transcription factor TAZ, resulting in committed monopotent osteoprogenitor cells with extensive proliferative capacity. Differentiation of the mature osteoblast is ultimately regulated by transcription factors, including Cbfa-1/Runx-2 and osterix. A diagramatic schema of the osteoblast lineage and its features is shown in Fig. 2.

The effects of growth factors on osteoblast differentiation


Huge advances have been made in the understanding of cellular and molecular control of bone formation in the past decade. The establishment of in vitro models of osteoblast differentiation and formation (11) have been essential for determining the effects of specic growth factors and growth factor-induced transcription factors on osteogenesis. These models include the use of primary cell cultures and established cell lines from a range of different species and

with distinct properties (98). Interpretation of the large amount of data from these in vitro models can sometimes be difcult owing to the variations seen in different model systems and the fact that cells may show different responses according to the precise stage of target cells through the lineage at different stages of differentiation. For example, as reviewed below, broblast growth factor-2 treatment of osteoprogenitor cells in vitro results in a mitotic response but suppression of the expression of osteoblast differentiation markers, but paradoxically, when applied in vivo, may result in an overall stimulation of bone formation. This can be explained by the suggestion that broblast growth factor-2 results in the expansion of relatively undifferentiated cells, which is seen as a suppression of differentiation in vitro but ultimately results in increased mature osteoblast numbers in vivo. Tissue engineering aims to produce tissues that are both structurally and functionally identical to the original tissues they are replacing, and strategies for growing bone therapeutically are emerging based on the knowledge of the physiological role of different

52

Effects of growth factors and cytokines on osteoblast differentiation

signaling molecules in the bone-forming process. The potential therapeutic application of growth factors in bone tissue engineering is considered further below. Bone formation is directed by the co-ordinated expression of many molecules, including growth factors, BMPs and specic transcription factors, which utilize developmentally derived signals to induce cellular and molecular stimuli to guide cellular commitment and differentiation in the proper spatial and temporal sequence. It is envisaged that future periodontal tissue engineering may involve the use of appropriate gene expression systems for controlled delivery of regenerative growth factors within the appropriate biological scaffold. Growth factors are soluble proteins that act as signaling agents for cells and inuence critical functions, such as cell division, matrix synthesis and tissue differentiation, by receptorligand binding. Results of experimental studies have established that growth factors play many important roles in bone formation and bone repair. Within the periodontal environment, growth factors found in bone, cementum and healing tissues include transforming growth factor-b, basic broblast growth factor, insulin-like growth factors, platelet-derived growth factor and BMPs. Recently, with the advent of recombinant growth factor proteins there has been considerable interest in their use as therapeutic agents in the treatment of bone disorders (including periodontal disease), and as growth factors become available as therapeutic agents, it is essential that we understand their biological characteristics and biological potential for periodontal tissue engineering. During bone formation, a number of candidate growth factors and their respective signaling pathways have been delineated, although there is a paucity of understanding of the complexities in the way that these growth factors interact to regulate bone formation and repair. It seems biologically sensible that there is involvement of multiple bioactive factors in osteoblast development and bone formation, with factors acting in a sequential manner. A number of growth factors, and their downstream molecular targets, have been characterized during osteoblast differentiation. These ndings suggest that at least some of the growth factors reviewed in this article, or their genetic effectors, may be potential therapeutic targets for regenerating bone. Growth factors that are known to affect osteogenic cells include transforming growth factor-b, broblast growth factors, platelet-derived growth factor and insulin-like growth factor.

Transforming growth factor-b


Transforming growth factor-b belongs to a large superfamily of related proteins that also includes BMPs, growth and differentiation factors, activins, inhibitins and anti-Mullerian hormone. All members play important roles in regulating cell proliferation and differentiation and the production of extracellular matrix. There are ve isoforms of transforming growth factor-b (transforming growth factor-b1 to transforming growth factor-b5). Most cells synthesize and respond to transforming growth factor-b, but high levels are found in bone, platelets and cartilage. Transforming growth factor-b1 is the most abundant isoform at the protein level (for a recent comprehensive review see Janssens et al. (111)). The activation of transforming growth factor-b is highly regulated, and once activated it interacts with transmembrane serine/threonine kinase receptors. Pivitol genetic studies identied that intracellular transforming growth factor-b signaling is mediated through mothers against dpp (Mad) in Drosophila melanogaster and subsequently the homologous genes in vertebrates (Smads) have been identied [reviewed in Massague and Chen (158)]. Although Smads are the key mediators in the transforming growth factor-b signaling pathway, BMPs have also been shown to signal through the Ras/mitogen-activated protein kinase (MAPK)/activator-protein-1 (AP-1) pathway [for review see Nohe et al. (176)]. During the early stages of bone formation, the action of transforming growth factor-b is to recruit and stimulate osteoprogenitor cells to proliferate, providing a pool of early osteoblasts (36, 206). In contrast, during later phases of osteoblast differentiation, transforming growth factor-b blocks differentiation and mineralization (154). These effects appear to be highly dependent on bone cell source, dose applied and the local environment, which may be a result of the inhibition of DNA synthesis at high transforming growth factor-b concentrations (35). Additionally, transforming growth factor-b inhibits the expression of the Runx2 and osteocalcin genes, whose expression is controlled by Cbfa1/ Runx2 in osteoblast-like cell lines, and this was found to be mediated by Smad3 (5). Transforming growth factor-b interacts with a range of other growth factors in bone with a resulting complex response. Further work is needed to clarify the role of transforming growth factor-b during periodontal regeneration and wound healing and to determine the inter-relationship between transforming growth factor-b and other growth factors that

53

Hughes et al.

have effects during different stages of osteoblast differentiation.

Bone morphogenetic proteins


Bone morphogenetic proteins are secreted signaling molecules which have a variety of functions during development (92) and cell differentiation (266). They were discovered for their remarkable ability to induce cartilage and bone formation from nonskeletal mesenchymal cells (252, 253) by recapitulating the entire sequence of events occurring during endochondral ossication. They have been subsequently cloned, sequenced and recombinant proteins manufactured (219, 268). These recombinant proteins have been the subject of many studies in vivo and in vitro and it is likely that these recombinant proteins will be used therapeutically in the near future for a range of applications in orthopedics, craniofacial surgery and dentistry (267). Like transforming growth factor-b, BMPs are expressed during embryonic development as well as into adulthood. Bone morphogenetic proteins are synthesized as a precursor molecule, and the active homodimeric or heterodimeric forms of BMP follow release of the C terminal by proteolysis (268). More than 20 BMP-related proteins have been identied, and there is diversity of biological activity as each BMP binds to their receptor with different afnities. Bone morphogenetic proteins bind to two distinct type I and II serine/threonine kinase receptors, both of which are required for signal transduction, although the type I receptor determines specicity of intracellular signals. Once activated, signals are transmitted intracellularly via Smaddependent and Smad-independent pathways [for review see Miyazono et al. (161)]. Among the BMP family, BMP-2, BMP-4 and BMP-7 have key roles during osteoblast commitment and differentiation. The primary effect of BMPs is on the pluripotent cells that are capable of differentiating into other mesenchymal cell types (10, 119, 258), and BMP-2 can direct these cells to commit to an osteoblastic pathway, as can BMP-4 and BMP-6 (102). Bone morphogenetic proteins can also increase the differentiation of committed cells to the osteoblast lineage, with the formation of bone nodules and expression of markers of the mature osteoblast phenotype (39, 102). Gene-expression studies, directly delivering an adenovirus with BMP-7 to gingival or dermal broblasts, leads to a robust osteogenic response (61). In recent years there has been an explosion of interest in potential molecular targets of BMPs, both

from the basic molecular pathway and potential therapeutic perspectives. This has given considerable insight into the genetic cascade leading to bone formation. Among these specic targets are the transcription factors Cbfa1/Runx2 (55), osterix (170) and TAZ (95). Bone morphogenetic proteins can upregulate Cbfa1/Runx2 under certain conditions during osteoblast differentiation; therefore, this is a candidate downstream target of BMPs, although Smad complexes can also directly interact and activate target genes independently of Cbfa1/Runx2 (115). Evidence is also emerging to suggest that some aspects of BMP-2-induced differentiation may be mediated through Wnt/b-catenin signaling, although co-operation between b-catenin and Smads are needed to activate late osteoblast gene expression (13). b-catenin is the classical effector of Wnt protein signaling, suggesting a role for this group of proteins in BMP-2 signal transduction.

Fibroblast growth factor


The broblast growth factors are a family of structurally related polypeptides that are known to play a critical role in angiogenesis and mesenchymal cell mitogenesis. To mediate their range of effects, broblast growth factor proteins signal via membrane-spanning tyrosine kinases (FGFR) and there are a wide variety of mechanisms for receptor regulation and availability. Mutations in these receptors are associated with abnormalities in ossication and activating mutations in FGFR2 cause several craniosynostosis syndromes by affecting the proliferation and differentiation of osteoblasts (185), highlighting a key role for these molecules in the control of bone formation. In normal adult tissues, the most abundant proteins are broblast growth factor-1 and broblast growth factor-2. Fibroblast growth factor-2 is expressed by osteoblasts and is generally more potent than broblast growth factor-1 (29), although the expression of other broblast growth factors are not nearly as ubiquitous. Although broblast growth factor signaling has been implicated in bone development, studies on null mutant mice have not yet fully shown the role of this family in skeletal development (166). Fibroblast growth factor-1 and broblast growth factor-2 in vitro stimulate osteoblast proliferation (in calvarial cells, ROS 17/2.8 and MC3T3-E1) but do not increase collagen production or alkaline phosphatase in differentiated osteoblasts (28, 106, 207), although these effects may be differentiation stage-specic as constitutive broblast growth factor signaling inhibits

54

Effects of growth factors and cytokines on osteoblast differentiation

osteoblastic differentiation (49) and dramatically increases apoptosis when cells are exposed to differentiating conditions (157). Fibroblast growth factors are strongly mitogenic to bone marrow stromal cells and are able to maintain the self-renewal of these cells in culture (135). It seems that short-term treatment with broblast growth factor, followed by its removal from culture, has an overall stimulatory effect on osteoblasts, and these growth factors may have potential as an adjunctive agent to increase bone formation. Their general effects in vitro are consistent with them acting mainly to stimulate proliferation in immature cells (whilst concomitantly inhibiting differentiation), resulting in expansion of the osteoblast progenitor pool.

Platelet-derived growth factor


Platelet-derived growth factor is secreted by platelets during the early phases of fracture healing, but its presence has been found in various tissues, including bone. Owing to its expression by a range of tissues, it is thought to have both systemic and local actions. Platelet-derived growth factor is composed of two polypeptide chains and can exist in three different isoforms of two gene products (AA,BB,AB) and these bind to two separate a and b receptors. Platelet-derived growth factor is a powerful mitogen for connective tissue cells, and although it can stimulate, and is synthesized by, mesenchymal cells and osteoblast like cells (83), it does not have powerful bone-induction properties. Platelet-derived growth factor isoforms have a strong chemotactic effect on osteoblasts and other connective tissue cells (100, 101), and may act to recruit mesenchymal cells during bone development and remodeling. Platelet-derived growth factor may also have direct and indirect effects on bone resorption by the upregulation of collagenase transcription (215) and an increase in interleukin-6 (IL-6) expression (64) in osteoblasts. In addition to platelet-derived growth factor autoregulation (214) in osteoblasts, there is paracrine regulation by other growth factors, such as transforming growth factor-b (213). Overall, the main physiological actions for platelet-derived growth factor in bone regulation are as nonspecic mitogens, but their effects on cell chemotaxis and neovasculogenesis may be particularly important during would healing.

mone participates in the regulation of skeletal growth and triggers the release of insulin-like growth factor in target cells. The insulin-like growth factors are bound to binding proteins, adding another crucial tier to modulate the activity of insulin-like growth factor. Two insulin-like growth factors have been identied insulin-like growth factor-1 and insulin-like growth factor-2 both of which are found in high concentration in serum. In bone, whilst insulin-like growth factor-2 is more abundant, insulin-like growth factor-1 may be more potent, although this might be different both between and within species (208). The regulation of insulin-like growth factor is complex, and the growth hormone mode of action in skeletal cells is largely unknown. Of the major hormones that regulate the skeleton, all have signicant effects on skeletal insulin-like growth factor, as do many growth factors, such as BMP-2 (27), transforming growth factor- (181) and broblast growth factor (70). Insulin-like growth factors increase proliferation and play a major role in stimulating mature osteoblast function. As with other growth factors detailed in this section, the way that osteoblasts respond to insulin-like growth factor signals may well depend on both the differentiation status of the cell and cell type. At the molecular level, insulin-like growth factor-1 upregulates the osteoblast-associated transcription factor, osterix, but not Cbfa1/Runx2. In addition, insulin-like growth factor-1, in combination with BMP-2, acts synergistically on osterix expression (34). Although it is widely accepted that insulin-like growth factors have a dening role in bone remodeling, their actual role is still unclear and needs to be understood within the complex inter-relationships of the components of the insulin-like growth factor system that evidently occur in vivo. Overall, the evidence suggests that the major effects of insulin-like growth factors are to promote the late-stage differentiation and activity of osteoblasts.

Wnt signaling
There is increasing evidence for the role of Wnt signaling in the control of early stages of the osteoblast lineage. Wnts are a group of over 15 related extracellular signaling molecules that show ligand binding with their receptor, frizzled, and co-receptors LRP5/6. Ligand binding sets off a series of intracellular reactions, resulting in stabilization of the protein b-catenin (200). This is then translated to the nucleus, where it acts as a co-factor with other DNA-binding proteins to regulate gene transcription.

Insulin-like growth factors


Growth hormone and insulin-like growth factors play critical roles in skeletal development. Growth hor-

55

Hughes et al.

This Wnt/b-catenin signaling pathway is also known as the canonical Wnt signaling pathway. In addition, Wnt ligand binding can regulate gene expression by noncanonical pathways without the involvement of b-catenin. The canonical Wnt signaling pathway has been shown to have important roles in the maintenance of self-renewal of stem cells in epidermal and hematopoietic cells, and there is increasing evidence for its role in the regulation of bone formation (31, 200, 262). First, it has been shown that canonical Wnt signaling can directly promote osteogenesis through actions on the Cbfa-1/Runx-2 gene (72). Second, genetic defects and polymorphisms of the Wnt co-receptor, LRP5, are associated with an osteoporotic phenotype (14, 129). The relationship between Wnt signaling and other growth factormediated osteoblast effects is, to date, confusing, with evidence for Wnt signaling acting through the induction of BMPs, and, conversely, BMPs acting through the induction of Wnt signaling in mesenchymal cells (13, 199, 265). At present there is little or no information about the role of Wnt signaling in local bone formation during wound healing, although it is likely that this signaling pathway may play an important role in the early events of local bone formation.

BMP expression by direct interaction with bmp gene promoters (121).

Enamel matrix derivatives


Enamel matrix derivatives (EMD) are the major component of commercially available Emdogain and are discussed here in order to compare information on their biological activity with that presented for growth factors above. The main biological effects of EMDs have been attributed to their predominant protein, amelogenin (37), with the remaining fraction comprising less-characterized factors. Amelogenin is not a classic growth factor, but rather a cell-adhesion matrix-bound protein, and specic amelogenin gene products are thought to have activity as epithelialmesenchymal signaling molecules. In addition, there is evidence that the alternate splice variant of the amelogenin gene, leucine-rich amelogenin peptide may also have direct signaling activities on cementoblasts, and, thus at least by implication, osteoblasts (20). Enamel matrix derivative has been used clinically for periodontal regeneration, and its therapeutic effectiveness has been variously attributed to amelogenin, nonamelogenin enamel matrix proteins and growth factors. While EMDs may induce periodontal regeneration, the precise mechanism of this is not known. In vitro studies show that EMDs can increase matrix production (86), proliferation and bone nodule formation of periodontal ligament cell cultures (73) and the differentiation of human and murine osteoblast cell lines (221), with the stimulation of phenotypic bone markers in some osteoblast cell lines (270). In contrast, some studies show no effect of EMDs on osteoblastic differentiation, although other growth factors were stimulated (183). In addition, noncommercial EMDs have been shown to contain both transforming growth factor-b and BMPlike growth factors (233). The osteogenic activity of EMDs may be mediated by the induction of BMP-like molecules, as EMDs induce Cbfa1/Runx2 expression and the phosphorylation of Smad1, both of which can be blocked by the BMP inhibitor, noggin (237). The search for downstream target genes has also revealed EMD response elements in the rat bone sialoprotein gene promoter that may mediate the effects of EMDs on bone sialoprotein gene transcription (223). A cDNA microarray study examining EMD-mediated changes in gene expression in periodontal ligament cells in vitro has reported the downregulation of genes involved in the early inammatory phases of wound healing, while simulta-

Other growth factors


There are many other growth factors that can interact with cells of the osteoblast lineage and may play a role in the local regulation of bone formation. For example, osteoprogenitor cells are responsive to epidermal growth factor stimulation, although epidermal growth factor receptors may be downregulated in mature osteoblastic cells (48). Treatment of cell cultures in vitro with epidermal growth factor results in increased cell proliferation and suppression of bone module formation (7). Epidermal growth factor utilizes many of the signal transduction pathways in common with platelet-derived growth factor although, using a proteomic approach, subtle differences in intracellular signaling have recently been reported in these two signaling mechanisms (137). Sonic hedgehog (Shh) is another growth factor-like protein which binds to a specic cell-surface receptor, patched (Ptc). Shh can induce osteoblastic commitment in cultured multipotent mesenchymal stem cells and promote osteoblast differentiation (126, 230, 271). These actions can be blocked with BMP antagonists, suggesting that Shh is an upstream regulator of BMP production and the Shh-activated intracellular signaling molecule, Gli, can upregulate

56

Effects of growth factors and cytokines on osteoblast differentiation

Fig. 3. Simplied schematic diagram showing the main stages of the osteoblast lineage where different growth factors may act. BMP, bone morphogenetic protein; IGF, insulin-like growth factor; FGF, broblast growth factor;

PDGF, platelet-derived growth factor; Shh, Sonic hedgehog; TGF-b, transforming growth factor-b; Wnts, a group of >15 related extracellular signaling molecules.

neously upregulating genes encoding growth and repair-promoting molecules (193). The in vitro treatment of cementoblasts with EMDs was found to decrease osteocalcin expression and to increase osteopontin expression (249). Overall, the data support the positive effect of EMDs on osteoblast differentiation, although further studies are needed to clarify which molecules in EMDs stimulate osteogenesis and to dene their precise modes of action.

outcome of these cellular responses may be both anabolic and catabolic. This section will provide an insight into the capacity of the major pro-inammatory cytokines to regulate osteoblast differentiation and function. Finally, the coupling of the osteoblast to the regulation of bone resorption will be introduced.

Interleukin-1
Interleukin-1 is a multipotent cytokine comprising two individual peptides, namely IL-1a and IL-1b, which exhibit similar biological activities (52). Both hematopoietic and mesenchymal/osteoblastic cells can produce IL-1 (89). There are two IL-1 receptors, namely type-I and type-II. All cellular responses elicited by IL-1 are known to be mediated through the type-I receptor (227), whereas type-II acts as a nonsignaling decoy receptor for the cytokine (41). Interleukin-1 also has a natural inhibitor, termed IL-1 receptor antagonist (IL-1ra). This binds to, but does not activate, IL-1 receptors (88), thus blocking the biological activity of the cytokine (9). The signaling events downsteam of the IL-1 receptor are mediated by IL-1 receptor-associated kinase (IRAK), and include the recruitment of phosphatidylinositol 3-kinase (PI3K), phosphorylation of MAPKs, and ultimately activation of the transcription factor, nuclear factor kappa B (NF-jB) (12). In the context of bone, IL-1 is known to regulate both resorption (149) and formation (25). However, the outcomes of different studies on the effects of IL-1 on osteoblast function are rather divergent. On the one

Summary
Considerable advances have been made in describing the regulation of osteoblasts by growth factors and in identifying some of the molecular mechanisms involved in these processes. Although the data are at times complex and sometimes contradictory, an overall pattern emerges of a carefully regulated, temporal sequence of co-ordinated growth factor expression, which can be modulated at any stage of the differentiation cycle. A schematic representation of this is shown in Fig. 3.

Effects of inammatory cytokines on osteoblast differentiation


Apart from responding to growth and differentiation factors, osteoblasts are also responsive to a number of locally acting cytokines, which are molecules that regulate cellular behavior of bone under situations of inammation, infection and wound healing. The

57

Hughes et al.

hand, both IL-1a and IL-1b have been shown to inhibit osteoblast proliferation and enhance bone formation, as demonstrated by enhanced alkaline phosphatase activity and bone nodule formation (50, 87, 179). On the other hand, depending on the differentiation stage of the cell, prolongation of the culture period and concentration of the cytokines, IL-1a and IL-1b, may stimulate osteoblast proliferation, including DNA and protein synthesis, and inhibit bone formation (25, 57, 202, 241), and inhibit osteocalcin and type I collagen production (231, 232). Interleukin-1 can also stimulate osteoblasts to produce other pro-inammatory cytokines, such as IL-6 (38, 110), IL-7 (261) and tumor necrosis factor-a (TNF-a) (259), or other inammatory mediators, such as prostaglandin E2 and nitric oxide (104, 105, 120, 197, 198).

extracellular signal-regulated protein kinase 1/2 MAPK, as well as the signal transducers and activators of transcription 1 and 3 (STAT-1 and STAT-3) (59, 65). Desensitization of IL-6 signaling is accomplished by endocytosis of the receptorligand complex, a process dependent on gp130. However, attenuation of IL-6-induced STAT-1 and STAT-3 expression, is independent of IL-6/IL-6R, or gp130 endocytosis (247, 248). Apart from the direct effects on osteoblast proliferation and differentiation, it is also possible that the IL-6/sIL-6R complex may indirectly regulate the expression of other factors known to control these cellular events. These include insulin-like growth factors (63, 62), BMPs (269), and a number of other paracrine and endocrine regulators of osteoblasts.

Tumor necrosis factor-a Interleukin-6


The IL-6 family of cytokines includes IL-6, IL-11, leukemia inhibitory factor, oncostatin M, cardiotrophin-1, and ciliary neurotrophic factor (216). Interleukin-6 is a pleiotropic cytokine crucial for the regulation of bone metabolism, reportedly more so than other members of the IL-6 family. The major source of this cytokine in bone is osteoblasts and stromal cells (94). Its involvement in bone remodeling is particularly evident in situations of increased bone turnover, rather than in physiological conditions (65, 93, 134, 156). It was initially demonstrated in vitro that IL-6 does not affect, or may slightly downregulate osteoblast differentiation (99, 110, 146, 147). These ndings were further strengthened by the absence of any pathological bone phenotype in IL-6 knockout mice (133). However, it is now established that the cellular expression of IL-6R, the cognate receptor to this cytokine, occurs at low levels. Therefore, the presence of the soluble receptor, sIL6R, an agonist for IL-6, is imperative for the activation of the downstream effects of this cytokine, in a process called trans-signaling (59, 65, 117). In the light of this requirement, studies on the osteoblast demonstrate that the IL-6/sIL-6R complex, IL-11, leukemia inhibitory factor and oncostatin M inhibit proliferation and stimulate osteoblastic cell differentiation via a cascade of intracellular events mediated by gp130, a signal-transducing receptor subunit (17, 58, 235). This molecule is more ubiquitously expressed than IL-6R (216). Binding of IL-6 to either its membrane-bound or its soluble receptor, results in homodimerization of gp130 and initiation of downstream signaling pathways, including the activation of the Janus Kinases and subsequently the Tumor necrosis factor-a is a member of the TNF family of inammatory cytokines that share common signaling pathways. It has a pivotal role in bone pathophysiology, because it has been shown to stimulate bone resorption and inhibit bone formation (19, 171). The major source of TNF-a is the T cell, and the cognate receptors to TNF-a are TNFR-1 (or p55) and TNFR-2 (or p75) (171). The former is required to initiate cell signaling, whereas the latter may contribute to the sensitivity of the cell response to the cytokine (77). Ligandreceptor binding activates TNF receptor-associated factors (TRAFs), mainly TRAF-1 and TRAF-2, in osteoblasts (16, 33), which in turn evoke a series of interconnected events, ultimately leading to the activation of NF-jB. In brief, NF-jB is a set of ve polypeptide transcription factors, called p50, p52, p65 (or Rel A), Rel B and c-Rel, which form heterodimers. The NF-jB heterodimers form an inactive complex with their I-jB inhibitors. Once phosphorylated by the I-jB kinases, I-jB is ubiquitinated and degraded in proteosomes. This results in the liberation and translocation of NF-jB into the nucleus, where it activates the transcription of a number of genes (21). Optimal induction of the respective target genes also requires phosphorylation of the NF-jB proteins (254). It should be noted that NF-jB knockout mice develop osteopetrosis as a result of a defect in osteoclast differentiation (108). Tumor necrosis factor-a is known to inhibit osteoblast function and differentiation. Suppression of the osteoblastic phenotype is characterized by the inhibition of cell proliferation and decreased production of a number of extracellular matrix components, such as type I collagen (82, 172, 210, 229). Indeed, genetic analyses have identied TNF-a-

58

Effects of growth factors and cytokines on osteoblast differentiation

responsive regions in the promoter of the respective genes (COL1A1 and COL1A2), indicating that NF-jB mediates the inhibition of collagen transcription (109, 116, 136, 163). Tumor necrosis factor-a also inhibits alkaline phosphatase and osteocalcin expression. However, the nature of osteocalcin inhibition is different from that of type I collagen, because this does not require binding of NF-jB to the promoter of the osteocalcin gene (173, 171). Apart from suppressing extracellular matrix production by osteoblasts, TNF-a also inhibits the differentiation of pre-osteoblasts as a result of the suppression of Cbfa1/Runx2 (1, 76) and osterix (171). In addition, TNF-a stimulates IL-6 production by osteoblasts (47, 110, 203) in an NF-jB-dependent mechanism (139). However, recent evidence suggests that other pathways such as p38 MAPK, and protein kinase C may also be involved in this event (138, 167). Tumor necrosis factor-a also induces prostaglandin E2 production by osteoblasts, an event at least partially mediated by nitric oxide (104).

Regulation of osteoclastogenesis and bone resorption by the osteoblast


In recent years it has become evident that osteoblasts have a global role in orchestrating the bone remodeling process. Their function is not restricted solely to bone formation, but it is now rmly established that they are responsible for initiating bone resorption. In cellular terms, apart from forming the mineral and organic extracellular compartment of bone, the osteoblast provides the essential and sufcient stimuli that control the behavior of the osteoclast, an event that occurs via cellcell interaction. The molecular determinants of this interaction are two factors produced by osteoblasts/stromal cells, namely M-CSF and RANKL, the former secreted and the latter mainly cell-membrane bound (244). M-CSF binds to c-Fms on the surface of osteoclast precursors, and this event enhances their proliferation and survival. RANKL, a member of the TNF ligand superfamily, is the factor that actually triggers the differentiation of osteoclast precursors, into mature osteoclasts and activates bone resorption, and is an absolute requirement for osteoclast formation (131, 140). Therefore, upon binding of RANKL to its cognate RANK receptor on the surface of the M-CSFtriggered osteoclast precursors, a number of downstream signaling pathways are activated, committing the cell to the osteoclast lineage. These are initially mediated by TRAF-6, subsequently leading to the activation of NF-jB, the AP-1 transcription factor

complex, and ultimately nuclear factor of activated T cells (NFAT) c1 (238, 245). The effects of RANKL can be blocked by the soluble nonsignaling decoy receptor, osteoprotegerin (OPG), a member of the TNFR superfamily with high homology to RANK (226). OPG is produced and secreted by osteoblasts and stromal cells, and by binding to RANKL it prevents RANKL/RANK interaction and consequently the downstream events described. RANKL-decient mice lack functional osteoclasts and develop osteopetrosis, whereas OPG-decient mice develop earlyonset osteoporosis, demonstrating the indispensable role of a balanced RANKL/OPG expression in physiological bone remodeling (23, 131). An increased RANKL/OPG expression ratio has been demonstrated in diseases involving inammation-induced bone loss, including periodontitis (148, 243, 246) and rheumatoid arthritis (60). Interleukin-1, IL-6 and TNF-a are all potent inducers of osteoclast differentiation and bone resorption. It is postulated that their catabolic effects on bone are mediated, to a great extent, through osteoblasts/stromal cells, and more specically through the regulation of RANKL and OPG expression in these cells (144, 148, 256). Indeed, it has been demonstrated that IL-6/sIL-6R, IL-11, leukemia inhibitory factor and oncostatin M increase RANKL and OPG expression in osteoblasts, in a manner dependent on gp130 and its downstream effector STAT-3 (177, 178, 191). Similarly, both IL-1 and TNFa induce RANKL expression, the former possibly being dependent on activation of STAT-3, whereas the latter is dependent on phosphorylation of p38 (47, 90, 259). OPG expression is also enhanced by these two cytokines (22, 255). Although both RANKL and OPG are upregulated in these systems, the overall outcome is an increased RANKL/OPG expression ratio, resulting in stimulation of osteoclastogenesis and bone resorption.

Other inammatory cytokines


A number of other cytokines have been reported to regulate bone cell behavior. Interleukin-7 (78) and IL-10 (71) have an inhibitory action on osteoblast differentiation. Interleukin-4 and IL-13 are known to inhibit osteoblast differentiation (251), and mice over-expressing IL-4 appear to have a decreased bone formation, owing to a reduction in the number and activity of bone-lining osteoblasts (145). Interleukin18 is structurally and functionally homologous to IL-1, but it employs differential signal-transducing pathways, preferentially activating p38 MAPK rather

59

Hughes et al.

than NF-jB (53, 143). It is produced by osteoblasts/ stromal cells, to which it acts as a mitogen, and is also known to inhibit osteoclast differentiation (43, 250). This may, in part, account for its capacity to stimulate osteoprotegerin, but not affect RANKL expression in osteoblasts/stromal cells (155). Interferon-c is a lymphokine produced by activated T cells, with a strong potency to inhibit osteoclastogenesis (80, 81, 239), It also inhibits osteoblast proliferation, as well as collagen and noncollagen protein synthesis (82, 172, 229). These effects appear to be dependent on nitric oxide production (103, 152, 153), rather than on prostaglandin E2 synthesis (229).

Summary
In summary, inammatory cytokines may have both anabolic and catabolic effects on osteoblast function, and consequently on bone physiology. However there is relatively little information on how these effects might also interact directly with growth factor pathways, but these interactions are likely to be very important. Studies have shown that both IL-1 and TNF-a may inuence the effects of BMPs on osteoblasts, but the nature of these interactions have not been explored at a mechanistic level (169, 240). Therefore, any tissue engineering approach targeting osteoblasts for bone regeneration must take into consideration the putative cellular responses elicited by cytokines upon them during inammation. This should be a concern, not only in the case of exogenously administered cytokines as a direct treatment strategy. It is highly probable that several other primary molecular agents may induce secondary inammatory responses by the target cells, inuencing, in turn, the cellular dynamics of bone in an autocrine or paracrine manner. This issue is particularly important in the instance of catabolic effects, which may occur either as inhibition of bone formation, or, most reportedly, as enhancement of bone resorption. Tackling efciently the potential sideeffects of these cytokines may help to ensure that the bone-remodeling balance would tend favorably toward bone formation, ultimately resulting clinically in bone regeneration.

in vitro or, on some occasions, from transgenic and null mutant (knockout) mouse models. These methodologies are extremely powerful for dissecting out specic signaling pathways as it is possible to control many of the extrinsic variables, thus facilitating the study of, for example, the role of individual specic molecules and signaling pathways. However, cells in vivo are exposed to a wide range of these signaling pathways simultaneously, and it is the total sum of these interactions which results in the carefully co-ordinated control of osteoblast function that is seen in vivo. Although there are some data on how different pathways may interact during the co-ordinated regulation of bone growth, at the present time it is only possible to provide examples of such interactions, rather than give a complete picture of the complex interactions in cytokine signaling pathways during bone growth. There are a number of mechanisms which can regulate different growth factor pathways. Different signaling pathways can also interact with each other in order to ne tune the nal observed outcome. These mechanisms are listed below. the regulation of growth factor activity by the secretion of soluble antagonists which are often regulated in both autocrine and paracrine fashions. the regulation of receptor expression on specic cells the interaction between intracellular signaling mechanisms in both antagonistic and synergistic ways.

Growth factor antagonists and binding proteins


Growth factors may bind to common proteins, such as a-2 macroglobulin, and be sequestered, for example, in matrices. Bone matrix is a rich source of many growth factors, including transforming growth factor-b and platelet-derived growth factor (45). In addition, many growth factors also appear to have specic binding proteins, which bind with their target factor with high afnity to inactivate them, or in some cases to increase ligand binding. Regulation of expression of growth factor antagonists and the balance of factor to antagonist is a clear mechanism for growth factors to control each others function in a sequential cascade, such as that described above. The activity of BMPs is regulated by a number of inhibitor proteins, including noggin, gremlin, follistatin and sclerostin, which bind to BMPs with high afnity in order to inactivate them (30). In addition, a novel BMP inhibitor, ectodin, has recently been

Regulation and interaction of growth factor pathways


Most of the information regarding the mechanisms of action of growth factors and cytokines on their target cells have been derived from studies carried out

60

Effects of growth factors and cytokines on osteoblast differentiation

described, whose expression is reportedly restricted to ectodermal-derived tissues in regulating epithelialmesenchymal interactions that are mediated by BMPs (142). The important actions that these inhibitors may have are suggested by a number of observations. First, mutations in the SOST gene (which codes for sclerostin) result in the rare bone disease, sclerosteosis, which is characterized by increased bone mass (15). Second, the overexpression of noggin in transgenic mice results in blocking the production of BMP-4-induced heterotopic bone formation (79). Third, evidence suggests that BMP-4-induced expression of noggin and gremlin is dysfunctional in patients with brodysplasia ossicans progressiva, a multilating condition where heterotopic bone forms in subdermal tissues (3). Finally, normal expression of ectodin in mice blocks enamel formation on one surface of the mouse incisor, and mice lacking ectodin have a number of defects in tooth formation, including an altered pattern of cusp formation and the formation of extra teeth (118, 142). Distinct BMP inhibitors may have tissue-specic activities and may also interact with different BMPs. For example, sclerostin expression is largely restricted to bone cells, particularly osteocytes, and may be involved in the osteocyte-mediated regulation of bone mass (264). In general, the expression of BMP antagonists is transcriptionally regulated by BMP signaling, giving rise to a negative feedback loop to regulate BMP signaling (30, 180). In addition, other pathways may regulate the expression of BMPs. In the case of ectodin, broblast growth factor and Shh signaling downregulates its expression, providing a mechanism whereby other growth factor signaling pathways may effectively upregulate BMP responses (142). The insulin-like growth factors are regulated by a series of at least six different binding proteins (insulin-like growth factor-binding proteins). As with other antagonists, insulin-like growth factor-binding proteins bind to the insulin-like growth factors with high afnity and typically may inactivate activity by preventing ligand binding. However, it is known that insulin-like growth factor-binding proteins may also act to mediate or regulate the ligand binding of insulin-like growth factors to their receptors on some occasions (26, 160). Insulin-like growth factors have been shown to differentially regulate insulin-like growth factor-binding protein gene expression (69). Insulin-like growth factor-binding proteins may also be regulated by a number of other osteotropic growth factors, including BMPs and transforming growth factor-b (68, 107, 160).

Fibroblast growth factors have complex interactions with a range of proteins and matrix molecules that regulate their function. First, it is known that broblast growth factors are normally matrix bound to heparan sulphate-containing proteoglycans and that heparan sulphate-containing proteoglycan binding is required for normal broblast growth factor ligandreceptor interactions (217). Alternatively spliced broblast growth factor receptor genes can give rise to broblast growth factor soluble receptors, which inactivate broblast growth factor by preventing normal receptorligand binding. Finally, there are additional broblast growth factorbinding proteins that also interact with broblast growth factors to control their function (141). The presence of these complex different levels of regulation together suggest a nely balanced set of mechanisms to control physiological broblast growth factor activity, although the specic functional signicance of broblast growth factor antagonists remains to be determined in bone. Transforming growth factor-b is also regulated closely by its association with binding proteins. Transforming growth factor-b is secreted in an inactive form bound to a specic binding peptide known as latency associated peptide. This latent transforming growth factor-b complex is then normally bound to one of at least four latent transforming growth factor-b-binding proteins (LTBPs) (182, 213). Latent transforming growth factor-b-binding proteins are extracellular matrix proteins found in bone matrix, and may be responsible for the large amounts of latent transforming growth factor-b sequestered in bone matrix. They may act as a delivery and release vehicle for transforming growth factor-b, and active transforming growth factor-b can be released from the complex by the enzymatic action of the plasminogen-activating system (6). In addition, a number of other proteins, including matrix metalloproteinases and thrombospondin, can release active transforming growth factor-b from latency associated peptide. The importance of these proteins is supported by the reports that LTBP-2 null mice die in utero at around stage e3.5; LTBP-3 knockout mice have a range of bone and craniofacial defects associated with a low turnover of bone (46). In contrast to most other growth factors, specic binding proteins have not been described for platelet-derived growth factor. However, platelet-derived growth factor binds to the matrix protein osteonectin (SPARC) and this may promote sequestering of platelet-derived growth factor in bone matrix and other sites (196).

61

Hughes et al.

Overall, it is clear that the production of growth factor-binding proteins is a mechanism whereby the activity of these proteins can be carefully controlled, and represents one level of regulation allowing careful cross-talk between different factors in a carefully co-ordinated way.

Interaction of growth factor intracellular signaling pathways


There are a number of examples described where different growth factor pathways regulate each other during intracellular signaling. Of particular interest, studies have demonstrated the regulation of SMAD signaling following BMP stimulation by additional growth factor signaling pathways (161). Recent results have demonstrated the downregulation of BMP-activated SMAD signaling by both transforming growth factor-b, via stimulating the expression of inhibitory SMAD (154), and by extracellular signalregulated protein kinase and PI3 kinase pathways, mediated by insulin-like growth factor-1 and other growth factor signals (186). These observations suggest many mechanisms whereby different growth factors regulate responsiveness to other signaling pathways. In this way it is possible that specic growth factor expression and signaling activity can be very delicately regulated to balance proliferation and differentiation events within cells of the osteoblast lineage. Overall, it is clear that although different cytokines may have distinct regulatory functions, during the control of bone formation, the interaction of these different pathways can be exquisitely regulated at a number of points. These observations, taken together, are consistent with a model of cytokine signaling consisting of sequential and overlapping expressions of factors to regulate nal bone-forming outcomes.

Therapeutic application of growth factors for bone regeneration


The topic of growth factor-induced bone and periodontal regeneration has been discussed extensively in other recent reviews (74, 234, 267), and it is not the intention in this article to revisit the detail of these experiments. However, here we consider some of the basic issues surrounding the use of exogenously applied growth factors to modulate bone growth, particularly in the periodontal tissues, and

briey describe the general ndings of some of these studies. Given the limitations of currently used clinical techniques, it is unsurprising that there is great interest in the possible utility of recombinant growth factors for tissue regeneration therapies. From a theoretical standpoint, the pharmacological application of growth factors results in a local single dose of the factor, which may be present at a concentration thousands of times higher than that normally seen physiologically. The pharmacological effects of the factor may be distinct from its physiological activity, for example because the normal regulatory feedback mechanisms which control that factor are temporarily overwhelmed by such a large dose. It seems quite likely that therapeutic application of any one of a number of growth factors could perturb the normal bone-forming process in a positive way and, indeed, preclinical studies support this idea. For example, based on the knowledge of physiological function, discussed above, BMPs may increase osteoblastic stem cell recruitment and commitment, broblast growth factor may stimulate early progenitor cell number expansion, platelet-derived growth factor may also increase progenitor cell recruitment and proliferation, and insulin-like growth factors might act to increase nal osteoblast differentiation and function. Any one of these actions may result in the desired nal outcome of increased bone formation. With this in mind, it is likely that one of the main critical determinants of success for these treatments is achieving sufcient substantivity for the factor to provide a continual therapeutic dose over a lengthy period of time (122). Although there is little published data on the pharmacokinetics of growth factor delivery, one study demonstrated that delivery of platelet-derived growth factor with insulin-like growth factor-I in a collagen matrix had a half-life of <4 h, with complete loss of activity by 96 h (151). Thus, in order to address this issue a number of different delivery systems have been used for these applications, including collagen matrix (123, 212), cross-linked gelatin pellets (124), calcium phosphate ceramics (112, 174), and polylactic-acid (PLA)-based resorbable polymers (192). In a different approach to provide persisting growth factor delivery, somatic cell gene transfer of platelet-derived growth factor has also been tested in vitro and in animal models (8, 61, 114, 113, 272). This approach might prove particularly successful in providing a sustained active concentration of growth factor to the site being treated. However, at present, serious concerns about the safety of the

62

Effects of growth factors and cytokines on osteoblast differentiation

adenovirus vectors used for gene transfer, and other ethical and practical issues, will need to be overcome before this could be thought of as a viable clinically applicable technique.

Platelet-derived growth factor and periodontal regeneration


A number of studies have tested the potential for tissue regeneration stimulated by platelet-derived growth factor and also with platelet-derived growth factor combined with insulin-like growth factor-1 or dexamethasone (40, 97, 112, 150, 151, 211, 212). In general terms, these studies (carried out in animals) have demonstrated an enhanced rate and an increased total amount of regenerated tissue compared with controls. However, there is little evidence of the efcacy of insulin-like growth factor-1, either on its own or in combination with platelet-derived growth factor (75). Most interestingly, a large multicentre human study (involving 180 patients) of platelet-derived growth factor has recently been reported (175). In that study, recombinant human platelet-derived growth factor-BB, with a b-tricalcium phosphate carrier, demonstrated improved bone ll and enhanced rate of clinical attachment gain compared with carrier-only controls. This is probably the rst large human study to assess the role of recombinant growth factor therapy and these interesting results will need further evaluation over time (201).

growth factors (162, 242). There is also clinical interest in the application of platelet-rich plasma, an autologous preparation obtained from a patients own platelets, which is rich in a number of growth factors, including platelet-derived growth factor and trefoil factor family-b, and which has been applied clinically in both periodontal and implant applications (32, 184, 218, 220). However, disappointingly, to date there is very little proper scientic evidence which evaluates the efcacy of this procedure, and well-controlled clinical studies of platelet-rich plasma would be valuable in ascertaining whether this treatment has a similar potential, for example, as the pharmacological use of recombinant growth factors.

Summary
The explosion of knowledge and the understanding of the role of growth factors, their mechanisms of action and molecular signaling pathways, which have been reviewed in this article, suggest the potential for many novel therapeutic targets, not only for applying growth factors but also for the potential use of growth factor inhibitors or agents that target specic parts of the intracellular signaling pathways. There remains an enormous challenge to convert some of the knowledge from basic studies of bone cell physiology to therapeutically useful techniques for the future. We are optimistic that such novel approaches may result in real qualitative improvements in clinical outcomes over currently available techniques.

Other growth factors


Bone morphogenetic proteins have also been widely tested for their ability to be used pharmacologically to enhance new bone formation. Bone morphogenetic proteins can powerfully promote bone formation in critical-size segmental defects in animals and have shown some promise for their ability to regulate bone and periodontal regeneration (123, 125, 205, 204, 224, 263). As these studies are the subject of a further separate review in this volume, they will not be considered further here. The effects of broblast growth factor-2 on animal models of periodontal regeneration have also been tested in animal models. Results of these studies are again promising, with broblast growth factor-2 showing a stimulation of parameters of bone and periodontal regeneration in surgically created defects (96, 164, 165, 168, 236). Other growth factors to have been tested include transforming growth factor-b where initial results of studies were disappointing when compared with the effects of these other

References
1. Abbas S, Zhang YH, Clohisy JC, Abu-Amer Y. Tumor necrosis factor-alpha inhibits pre-osteoblast differentiation through its type-1 receptor. Cytokine 2003: 22: 3341. 2. Ahdjoudj S, Fromigue O, Marie PJ. Plasticity and regulation of human bone marrow stromal osteoprogenitor cells: potential implication in the treatment of age-related bone loss. Histol Histopathol 2004: 19: 151157. 3. Ahn J, Serrano de la Pena L, Shore EM, Kaplan FS. Paresis of a bone morphogenetic protein-antagonist response in a genetic disorder of heterotopic skeletogenesis. J Bone Joint Surg Am 2003: 85-A: 667674. 4. Allan EH, Ho PW, Umezawa A, Hata J, Makishima F, Gillespie MT, Martin TJ. Differentiation potential of a mouse bone marrow stromal cell line. J Cell Biochem 2003: 90: 158169. 5. Alliston T, Choy L, Ducy P, Karsenty G, Derynck R. TGFbeta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. Embo J 2001: 20: 22542272. 6. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell Sci 2003: 116: 217224.

63

Hughes et al.
7. Antosz ME, Bellows CG, Aubin JE. Effects of transforming growth factor and epidermal growth factor on cell proliferation and the formation of bone nodules in isolated fetal rat calvaria cells. J Cell Physiol 1989: 140: 386395. 8. Anusaksathien O, Webb SA, Jin QM, Giannobile WV. Platelet-derived growth factor gene delivery stimulates ex vivo gingival repair. Tissue Eng 2003: 9: 745756. 9. Arend WP, Welgus HG, Thompson RC, Eisenberg SP. Biological properties of recombinant human monocytederived interleukin 1 receptor antagonist. J Clin Invest 1990: 85: 16941697. 10. Asahina I, Sampath TK, Hauschka PV. Human osteogenic protein-1 induces chondroblastic, osteoblastic, and/or adipocytic differentiation of clonal murine target cells. Exp Cell Res 1996: 222: 3847. 11. Aubin JE. Advances in the osteoblast lineage. Biochem Cell Biol 1998: 76: 899910. 12. Auron PE. The interleukin 1 receptor: ligand interactions and signal transduction. Cytokine Growth Factor Rev 1998: 9: 221237. 13. Bain G, Muller T, Wang X, Papkoff J. Activated betacatenin induces osteoblast differentiation of C3H10T1/2 cells and participates in BMP2 mediated signal transduction. Biochem Biophys Res Commun 2003: 301: 8491. 14. Baldock PA, Eisman JA. Genetic determinants of bone mass. Curr Opin Rheumatol 2004: 16: 450456. 15. Balemans W, Van Hul W. Identication of the diseasecausing gene in sclerosteosis discovery of a novel bone anabolic target? J Musculoskelet Neuronal Interact 2004: 4: 139142. 16. Beck GR, Jr, Zerler B, Moran E. Gene array analysis of osteoblast differentiation. Cell Growth Differ 2001: 12: 6183. 17. Bellido T, Borba VZ, Roberson P, Manolagas SC. Activation of the Janus kinase/STAT (signal transducer and activator of transcription) signal transduction pathway by interleukin-6-type cytokines promotes osteoblast differentiation. Endocrinology 1997: 138: 36663676. 18. Beresford JN, Bennett JH, Devlin C, Leboy PS, Owen ME. Evidence for an inverse relationship between the differentiation of adipocytic and osteogenic cells in rat marrow stromal cell cultures. J Cell Sci 1992: 102: 341351. 19. Bertolini DR, Nedwin GE, Bringman TS, Smith DD, Mundy GR. Stimulation of bone resorption and inhibition of bone formation in vitro by human tumour necrosis factors. Nature 1986: 319: 516518. 20. Boabaid F, Gibson CW, Kuehl MA, Berry JE, Snead ML, Nociti FH Jr, Katchburian E, Somerman MJ. Leucine-rich amelogenin peptide: a candidate signaling molecule during cementogenesis. J Periodontol 2004: 75: 11261136. 21. Boyce BF, Xing L, Franzoso G, Siebenlist U. Required and nonessential functions of nuclear factor-kappa B in bone cells. Bone 1999: 25: 137139. 22. Brandstrom H, Jonsson KB, Vidal O, Ljunghall S, Ohlsson C, Ljunggren O. Tumor necrosis factor-alpha and -beta upregulate the levels of osteoprotegerin mRNA in human osteosarcoma MG-63 cells. Biochem Biophys Res Commun 1998: 248: 454457. 23. Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C, Scully S, Tan HL, Xu W, Lacey DL, Boyle WJ, Simonet WS. Osteoprotegerin-decient mice develop early onset osteoporosis and arterial calcication. Genes Dev 1998: 12: 12601268. Burger EH, Klein-Nulend J, Smit TH. Strain-derived canalicular uid ow regulates osteoclast activity in a remodelling osteon a proposal. J Biomech 2003: 36: 14531459. Canalis E. Interleukin-1 has independent effects on deoxyribonucleic acid and collagen synthesis in cultures of rat calvariae. Endocrinology 1986: 118: 7481. Canalis E. Insulin-like growth factors and osteoporosis. Bone 1997: 21: 215216. Canalis E, Gabbitas B. Bone morphogenetic protein 2 increases insulin-like growth factor I and II transcripts and polypeptide levels in bone cell cultures. J Bone Miner Res 1994: 9: 19992005. Canalis E, Raisz LG. Effect of broblast growth factor on cultured fetal rat calvaria. Metabolism 1980: 29: 108114. Canalis E, Centrella M, McCarthy T. Effects of basic broblast growth factor on bone formation in vitro. J Clin Invest 1988: 81: 15721577. Canalis E, Economides AN, Gazzerro E. Bone morphogenetic proteins, their antagonists, and the skeleton. Endocr Rev 2003: 24: 218235. Canalis E, Deregowski V, Pereira RC, Gazzerro E. Signals that determine the fate of osteoblastic cells. J Endocrinol Invest 2005: 28: 37. Carlson NE, Roach RB Jr. Platelet-rich plasma: clinical applications in dentistry. J Am Dent Assoc 2002: 133: 1383 1386. Carpentier I, Beyaert R. TRAF1 is a TNF inducible regulator of NF-kappaB activation. FEBS Lett 1999: 460: 246 250. Celil AB, Campbell PG, Datto M, Wang XF, Hollinger JO, Selvamurugan N, Kwok S, Alliston T, Reiss M, Partridge NC, Hassel S, Schmitt S, Hartung A, Roth M, Nohe A, Petersen N, Ehrlich M, Henis YI, Sebald W, Knaus P, Lee KS, Hong SH, Bae SC, Lai CF, Cheng SL. 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: 3135331359. Centrella M, McCarthy TL, Canalis E. Transforming growth factor beta is a bifunctional regulator of replication and collagen synthesis in osteoblast-enriched cell cultures from fetal rat bone. J Biol Chem 1987: 262: 2869 2874. Centrella M, Horowitz MC, Wozney JM, McCarthy TL. Transforming growth factor-beta gene family members and bone. Endocr Rev 1994: 15: 2739. Cerny R, Slaby I, Hammarstrom L, Wurtz T. A novel gene expressed in rat ameloblasts codes for proteins with cell binding domains. J Bone Miner Res 1996: 11: 883891. Chaudhary LR, Spelsberg TC, Riggs BL. Production of various cytokines by normal human osteoblast-like cells in response to interleukin-1 beta and tumor necrosis factor-alpha: lack of regulation by 17 beta-estradiol. Endocrinology 1992: 130: 25282534. Chen TL, Bates RL, Dudley A, Hammonds R Jr, Amento EP. Bone morphogenetic protein-2b stimulation of growth and osteogenic phenotypes in rat osteoblast-like cells: comparison with TGF-beta 1. J Bone Miner Res 1991: 6: 13871393.

24.

25.

26. 27.

28. 29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

64

Effects of growth factors and cytokines on osteoblast differentiation


40. Cho MI, Lin WL, Genco RJ. Platelet-derived growth factormodulated guided tissue regenerative therapy. J Periodontol 1995: 66: 522530. 41. Colotta F, Re F, Muzio M, Bertini R, Polentarutti N, Sironi M, Giri JG, Dower SK, Sims JE, Mantovani A. Interleukin-1 type II receptor: a decoy target for IL-1 that is regulated by IL-4. Science 1993: 261: 472475. 42. Conget PA, Allers C, Minguell JJ. Identication of a discrete population of human bone marrow-derived mesenchymal cells exhibiting properties of uncommitted progenitors. J Hematother Stem Cell Res 2001: 10: 749758. 43. Cornish J, Gillespie MT, Callon KE, Horwood NJ, Moseley JM, Reid IR. Interleukin-18 is a novel mitogen of osteogenic and chondrogenic cells. Endocrinology 2003: 144: 11941201. 44. de Crombrugghe B, Lefebvre V, Behringer RR, Bi W, Murakami S, Huang W. Transcriptional mechanisms of chondrocyte differentiation. Matrix Biol 2000: 19: 389 394. 45. Crookston KP, Webb DJ, Lamarre J, Gonias SL. Binding of platelet-derived growth factor-BB and transforming growth factor-beta 1 to alpha 2-macroglobulin in vitro and in vivo: comparison of receptor-recognized and nonrecognized alpha 2-macroglobulin conformations. Biochem J 1993: 293 (Pt 2): 443450. 46. Dabovic B, Levasseur R, Zambuto L, Chen Y, Karsenty G, Rifkin DB. Osteopetrosis-like phenotype in latent TGFbeta binding protein 3 decient mice. Bone 2005: 37: 2531. 47. Dai JC, He P, Chen X, Greeneld EM. TNFalpha and PTH utilize distinct mechanisms to induce IL-6 and RANKL expression with markedly different kinetics. Bone 2006: 38: 509520. 48. Davideau JLSCTIBA. EGF receptor expression in mineralized tissues: an in situ hybridization and immunocytochemical investigation in rat and human mandibles. Conn Tissue Res 1995: 32: 4753. 49. Debiais F, Hott M, Graulet AM, Marie PJ. The effects of broblast growth factor-2 on human neonatal calvaria osteoblastic cells are differentiation stage specic. J Bone Miner Res 1998: 13: 645654. 50. Dedhar S, Mitchell MD, Pierschbacher MD. The osteoblast-like differentiated phenotype of a variant of MG-63 osteosarcoma cell line correlated with altered adhesive properties. Conn Tissue Res 1989: 20: 4961. 51. Dennis JE, Carbillet JP, Caplan AI, Charbord P. The STRO1+ marrow cell population is multipotential. Cells Tissues Organs 2002: 170: 7382. 52. Dinarello CA. Interleukin-1 and its biologically related cytokines. Adv Immunol 1989: 44: 153205. 53. Dinarello CA. Interleukin-18. Methods 1999: 19: 121132. 54. Ducy P, Desbois C, Boyce B, Pinero G, Story B, Dunstan C, Smith E, Bonadio J, Goldstein S, Gundberg C, Bradley A, Karsenty G. Increased bone formation in osteocalcindecient mice. Nature 1996: 382: 448452. 55. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/ Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 1997: 89: 747754. 56. Ducy P, Zhang R, Geoffroy V, Ridall A, Karsenty G. Osf2/ Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 1997: 89: 677680. 57. Ellies LG, Aubin JE. Temporal sequence of interleukin 1 alpha-mediated stimulation and inhibition of bone formation by isolated fetal rat calvaria cells in vitro. Cytokine 1990: 2: 430437. Erices A, Conget P, Rojas C, Minguell JJ. Gp130 activation by soluble interleukin-6 receptor/interleukin-6 enhances osteoblastic differentiation of human bone marrowderived mesenchymal stem cells. Exp Cell Res 2002: 280: 2432. Ernst M, Jenkins BJ. Acquiring signalling specicity from the cytokine receptor gp130. Trends Genet 2004: 20: 2332. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature 2003: 423: 356361. Franceschi RT, Wang D, Krebsbach PH, Rutherford RB. Gene therapy for bone formation: in vitro and in vivo osteogenic activity of an adenovirus expressing BMP7. J Cell Biochem 2000: 78: 476486. Franchimont N, Gangji V, Durant D, Canalis E. Interleukin-6 with its soluble receptor enhances the expression of insulin-like growth factor-I in osteoblasts. Endocrinology 1997: 138: 52485255. Franchimont N, Durant D, Canalis E. Interleukin-6 and its soluble receptor regulate the expression of insulin-like growth factor binding protein-5 in osteoblast cultures. Endocrinology 1997: 138: 33803386. Franchimont N, Durant D, Rydziel S, Canalis E. Plateletderived growth factor induces interleukin-6 transcription in osteoblasts through the activator protein-1 complex and activating transcription factor-2. J Biol Chem 1999: 274: 67836789. Franchimont N, Wertz S, Malaise M. Interleukin-6: an osteotropic factor inuencing bone formation? Bone, 2005: 37: 601606. Friedenstein AJ. Precursor cells of mechanocytes. Int Rev Cytol 1976: 47: 327359. Friedenstein AJ, Chailalhyan RK, Gerasimov UV. Bone marrow osteogenic stem cells: In vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet 1987: 20: 263272. Gabbitas B, Canalis E. Bone morphogenetic protein-2 inhibits the synthesis of insulin-like growth factor-binding protein-5 in bone cell cultures. Endocrinology 1995: 136: 23972403. Gabbitas B, Canalis E. Insulin-like growth factors sustain insulin-like growth factor-binding protein-5 expression in osteoblasts. Am J Physiol 1998: 275: E222E228. Gangji V, Rydziel S, Gabbitas B, Canalis E. Insulin-like growth factor II promoter expression in cultured rodent osteoblasts and adult rat bone. Endocrinology 1998: 139: 22872292. Garcia-Lopez S, Meikle MC, Villanueva RE, Montano L, Masso F, Ramirez-Amador V, Bojalil R. Mechanical deformation inhibits IL-10 and stimulates IL-12 production by mouse calvarial osteoblasts in vitro. Arch Oral Biol 2005: 50: 449452. 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: 3313233140. Gestrelius S, Andersson C, Lidstrom D, Hammarstrom L, Somerman M. In vitro studies on periodontal ligament cells and enamel matrix derivative. J Clin Periodontol 1997: 24: 685692.

58.

59. 60. 61.

62.

63.

64.

65.

66. 67.

68.

69.

70.

71.

72.

73.

65

Hughes et al.
74. Giannobile WV, Somerman MJ. Growth and amelogeninlike factors in periodontal wound healing. A systematic review. Ann Periodontol 2003: 8: 193204. 75. Giannobile WV, Hernandez RA, Finkelman RD, Ryan S, Kiritsy CP, DAndrea M, Lynch SE. Comparative effects of platelet-derived growth factor-BB and insulin-like growth factor-I, individually and in combination, on periodontal regeneration in Macaca fascicularis. J Periodontal Res 1996: 31: 301312. 76. Gilbert L, He X, Farmer P, Rubin J, Drissi H, van Wijnen AJ, Lian JB, Stein GS, Nanes MS. Expression of the osteoblast differentiation factor RUNX2 (Cbfa1/AML3/Pebp2alpha A) is inhibited by tumor necrosis factor-alpha. J Biol Chem 2002: 277: 26952701. 77. Gilbert LC, Rubin J, Nanes MS. The p55 TNF receptor mediates TNF inhibition of osteoblast differentiation independently of apoptosis. Am J Physiol Endocrinol Metab 2005: 288: E1011E1018. 78. Giuliani N, Colla S, Morandi F, Lazzaretti M, Sala R, Bonomini S, Grano M, Colucci S, Svaldi M, Rizzoli V. Myeloma cells block RUNX2/CBFA1 activity in human bone marrow osteoblast progenitors and inhibit osteoblast formation and differentiation. Blood 2005: 106: 24722483. 79. Glaser DL, Economides AN, Wang L, Liu X, Kimble RD, Fandl JP, Wilson JM, Stahl N, Kaplan FS, Shore EM. In vivo somatic cell gene transfer of an engineered Noggin mutein prevents BMP4-induced heterotopic ossication. J Bone Joint Surg Am 2003. 23322342. 80. Gowen M, Mundy GR. Actions of recombinant interleukin 1, interleukin 2, and interferon-gamma on bone resorption in vitro. J Immunol 1986: 136: 24782482. 81. Gowen M, Nedwin GE, Mundy GR. Preferential inhibition of cytokine-stimulated bone resorption by recombinant interferon gamma. J Bone Miner Res 1986: 1: 469474. 82. Gowen M, MacDonald BR, Russell RG. Actions of recombinant human gamma-interferon and tumor necrosis factor alpha on the proliferation and osteoblastic characteristics of human trabecular bone cells in vitro. Arthritis Rheum 1988: 31: 15001507. 83. Graves DT, Valentin-Opran A, Delgado R, Valente AJ, Mundy G, Piche J. The potential role of platelet-derived growth factor as an autocrine or paracrine factor for human bone cells. Connect Tissue Res 1989: 23: 209 218. 84. Grigoriades AE, Heersche JNM, Aubin JE. Differentiation of muscle, fat, cartilage and bone from progenitor cells present in a bone derived clonal cell population: Effects of dexamethasone. J Cell Biol 1988: 106: 21392151. 85. Gronthos S, Graves SE, Ohta S, Simmons PJ. The STRO-1+ fraction of adult human bone marrow contains the osteogenic precursors. Blood 1994: 84: 41644173. 86. Haase HR, Bartold PM. Enamel matrix derivative induces matrix synthesis by cultured human periodontal broblast cells. J Periodontol 2001: 72: 341348. 87. Hanazawa S, Ohmori Y, Amano S, Hirose K, Miyoshi T, Kumegawa M, Kitano S. Human puried interleukin-1 inhibits DNA synthesis and cell growth of osteoblastic cell line (MC3T3-E1), but enhances alkaline phosphatase activity in the cells. FEBS Letts 1986: 203: 279284. 88. Hannum CH, Wilcox CJ, Arend WP, Joslin FG, Dripps DJ, Heimdal PL, Armes LG, Sommer A, Eisenberg SP, Thompson RC. Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor. Nature 1990: 343: 336 340. Haynes DR, Atkins GJ, Loric M, Crotti TN, Geary SM, Findlay DM. Bidirectional signaling between stromal and hemopoietic cells regulates interleukin-1 expression during human osteoclast formation. Bone 1999: 25: 269278. Hofbauer LC, Lacey DL, Dunstan CR, Spelsberg TC, Riggs BL, Khosla S. Interleukin-1beta and tumor necrosis factoralpha, but not interleukin-6, stimulate osteoprotegerin ligand gene expression in human osteoblastic cells. Bone 1999: 25: 255259. Hoffmann HM, Catron KM, van Wijnen AJ, McCabe LR, Lian JB, Stein GS, Stein JL. Transcriptional control of the tissue-specic, developmentally regulated osteocalcin gene requires a binding motif for the Msx family of homeodomain proteins. Proc Natl Acad Sci U S A 1994: 91: 1288712891. Hogan B. Bone morphogenetic proteins in development. Curr Opin Genet Dev 1996: 6: 432438. Holt I, Cooper RG, Denton J, Meager A, Hopkins SJ. Cytokine inter-relationships and their association with disease activity in arthritis. Br J Rheumatol 1992: 31: 725733. Holt I, Davie MW, Marshall MJ. Osteoclasts are not the major source of interleukin-6 in mouse parietal bones. Bone 1996: 18: 221226. 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: 10741078. Hosokawa R, Kikuzaki K, Kimoto T, Matsuura T, Chiba D, Wadamoto M, Sato Y, Maeda M, Sano A, Akagawa Y. Controlled local application of basic broblast growth factor (FGF-2) accelerates the healing of GBR. An experimental study in beagle dogs. Clin Oral Implants Res 2000: 11: 345353. Howell TH, Fiorellini JP, Paquette DW, Offenbacher S, Giannobile WV, Lynch SE. A phase I/II clinical trial to evaluate a combination of recombinant human plateletderived growth factor-BB and recombinant human insulin-like growth factor-I in patients with periodontal disease. J Periodontol 1997: 68: 11861193. Hughes FJ, Aubin JE. Culture of cells of the osteoblast lineage. In: Arnett TR, Henderson B, editors. Methods in Bone Biology. London: Chapman Hall, 1998: 149. Hughes FJ, Howells GL. Interleukin-6 inhibits bone formation in vitro. Bone Miner 1993: 21: 2128. Hughes FJ, Mc Culloch CAG. Quantication of chemotactic response of quiescent and proliferating broblasts in Boyden chambers by computer-assisted image analysis. J Histochem Cytochem 1991: 39: 243246. Hughes FJ, Aubin JE, Heersche JNM. Differential chemotactic responses of different populations of fetal rat calvaria cells to platelet-derived growth factor and transforming growth factorb. Bone Miner 1992: 19: 6373. Hughes FJ, Collyer J, Staneld M, Goodman SA. The effects of bone morphogenetic protein-2, -4 and -6 on differentiation of rat osteoblast cells in vitro. Endocrinology 1995: 136: 26712677. Hughes FJ, Buttery L, Hukkanen M, Polak JM. Regulation of osteoblast activity by nitric oxide. In: Hukkanen M, Polak JM, Hughes SPF, editors. Nitric Oxide in Bone and

89.

90.

91.

92. 93.

94.

95.

96.

97.

98.

99. 100.

101.

102.

103.

66

Effects of growth factors and cytokines on osteoblast differentiation


Joint Disease. Cambridge: Cambridge University Press, 1998: 105115. Hughes FJ, Buttery LD, Hukkanen MV, ODonnell A, Maclouf J, Polak JM. Cytokine-induced prostaglandin E2 synthesis and cyclooxygenase-2 activity are regulated both by a nitric oxide-dependent and -independent mechanism in rat osteoblasts in vitro. J Biol Chem 1999: 274: 17761782. Hukkanen M, Hughes F, Buttery L, Gross S, Evans T, Seddon S, Riveros-Moreno V, MacIntyre I, Polak J. Cytokine-stimulated expression of inducible nitric oxide synthase by mouse, rat and human osteoblast-like cell and its functional role in osteoblast metabolic activity. Endocrinology 1995: 136: 54455453. Hurley MM, Abreu C, Harrison JR, Lichtler AC, Raisz LG, Kream BE. Basic broblast growth factor inhibits type I collagen gene expression in osteoblastic MC3T3-E1 cells. J Biol Chem 1993: 268: 55885593. Hwa V, Oh Y, Rosenfeld RG. Insulin-like growth factor binding protein-3 and -5 are regulated by transforming growth factor-beta and retinoic acid in the human prostate adenocarcinoma cell line PC-3. Endocrine 1997: 6: 235242. Iotsova V, Caamano J, Loy J, Yang Y, Lewin A, Bravo R. Osteopetrosis in mice lacking NF-kappaB1 and NF-kappaB2. Nat Med 1997: 3: 12851289. Iraburu MJ, Dominguez-Rosales JA, Fontana L, Auster A, Garcia-Trevijano ER, Covarrubias-Pinedo A, Rivas-Estilla AM, Greenwel P, Rojkind M. Tumor necrosis factor alpha downregulates expression of the alpha1(I) collagen gene in rat hepatic stellate cells through a p20C/EBPbeta- and C/EBPdelta-dependent mechanism. Hepatology 2000: 31: 10861093. Ishimi Y, Miyaura C, Jin CH, Akatsu T, Abe E, Nakamura Y, Yamaguchi A, Yoshiki S, Matsuda T, Hirano T, Kishimoto T, Suda T. IL-6 is produced by osteoblasts and induces bone resorption. J Immunol 1990: 145: 32973303. Janssens K, ten Dijke P, Janssens S, Van Hul W. Transforming growth factor-beta1 to the bone. Endocr Rev 2005: 26: 743774. Jensen SS, Broggini N, Weibrich G, Hjorting-Hansen E, Schenk R, Buser D. Bone regeneration in standardized bone defects with autografts or bone substitutes in combination with platelet concentrate: a histologic and histomorphometric study in the mandibles of minipigs. Int J Oral Maxillofac Implants 2005: 20: 703712. Jin QM, Anusaksathien O, Webb SA, Rutherford RB, Giannobile WV. Gene therapy of bone morphogenetic protein for periodontal tissue engineering. J Periodontol 2003: 74: 202213. Jin Q, Anusaksathien O, Webb SA, Printz MA, Giannobile WV. Engineering of tooth-supporting structures by delivery of PDGF gene therapy vectors. Mol Ther 2004: 9: 519 526. Jonk LJ, Itoh S, Heldin CH, ten Dijke P, Kruijer W. Identication and functional characterization of a Smad binding element (SBE) in the JunB promoter that acts as a transforming growth factor-beta, activin, and bone morphogenetic protein-inducible enhancer. J Biol Chem 1998: 273: 2114521152. Kahari VM, Chen YQ, Su MW, Ramirez F, Uitto J. Tumor necrosis factor-alpha and interferon-gamma suppress the activation of human type I collagen gene expression by transforming growth factor-beta 1. Evidence for two distinct mechanisms of inhibition at the transcriptional and posttranscriptional levels. J Clin Invest 1990: 86: 1489 1495. Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol 2003: 149: 138. Kassai Y, Munne P, Hotta Y, Penttila E, Kavanagh K, Ohbayashi N, Takada S, Thesleff I, Jernvall J, Itoh N. Regulation of mammalian tooth cusp patterning by ectodin. Science 2005: 309: 20672070. Katagiri T, Yamaguchi A, Ikeda T, Yoshiki S, Wozney JM, Rosen V, Wang EA, Tanaka H, Omura S, Suda T. The nonosteogenic mouse pluripotent cell line, C3H10T1/2, is induced to differentiate into osteoblastic cells by recombinant human bone morphogenetic protein-2. Biochem Biophys Res Comm 1990: 172: 295299. Kawaguchi H, Yavari R, Stover ML, Rowe DW, Raisz LG, Pilbeam CC. Measurement of interleukin-1 stimulated constitutive prostaglandin G/H synthase (cyclooxygenase) mRNA levels in osteoblastic MC3T3-E1 cells using competitive reverse transcriptase polymerase chain reaction. Endocrine Res 1994: 20: 219233. Kawai S, Sugiura T. Characterization of human bone morphogenetic protein (BMP)-4 and -7 gene promoters: activation of BMP promoters by Gli, a sonic hedgehog mediator. Bone 2001: 29: 5461. King GN. The importance of drug delivery to optimize the effects of bone morphogenetic proteins during periodontal regeneration. Curr Pharm Biotechnol 2001: 2: 131 142. King GN, King N, Cruchley AT, Wozney JM, Hughes FJ. Recombinant human bone morphogenetic protein-2 promotes wound healing in rat periodontal fenestration defects. J Dent Res 1997: 76: 14601470. King GN, King N, Hughes FJ. Effect of two delivery systems for recombinant human bone morphogenetic protein-2 on periodontal regeneration in vivo. J Periodontal Res 1998: 33: 226236. Kinoshita A, Oda S, Takahashi K, Yokota S, Ishikawa I. Periodontal regeneration by application of recombinant human bone morphogenetic protein-2 to horizontal circumferential defects created by experimental periodontitis in beagle dogs. J Periodontol 1997: 68: 103109. Kinto N, Iwamoto M, Enomoto-Iwamoto M, Noji S, Ohuchi H, Yoshioka H, Kataoka H, Wada Y, Yuhao G, Takahashi HE, Yoshiki S, Yamaguchi A. Fibroblasts expressing Sonic hedgehog induce osteoblast differentiation and ectopic bone formation. FEBS Lett 1997: 404: 319323. Klein-Nulend J, Semeins CM, Ajubi NE, Nijweide PJ, Burger EH. Pulsating uid ow increases nitric oxide (NO) synthesis by osteocytes but not periosteal broblasts correlation with prostaglandin upregulation. Biochem Biophys Res Comm 1995: 217: 640648. Knothe Tate ML, Steck R, Forwood MR, Niederer P. In vivo demonstration of load-induced uid ow in the rat tibia and its potential implications for processes associated with functional adaptation. J Exp Biol 2000: 203: 2737 2745. Koay MA, Brown MA. Genetic disorders of the LRP5-Wnt signalling pathway affecting the skeleton. Trends Mol Med 2005: 11: 129137.

104.

117.

118.

105.

119.

106.

120.

107.

108.

121.

109.

122.

123.

110.

124.

111.

112.

125.

126.

113.

114.

127.

115.

128.

116.

129.

67

Hughes et al.
130. 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: 755764. 131. Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, Morony S, Oliveira-dos-Santos AJ, Van G, Itie A, Khoo W, Wakeham A, Dunstan CR, Lacey DL, Mak TW, Boyle WJ, Penninger JM. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 1999: 397: 315323. 132. Konieczny SF, Emerson CP Jr. 5-Azacytidine induction of stable mesodermal stem cell lineages from 10T1/2 cells: evidence for regulatory genes controlling determination. Cell 1984: 38: 791800. 133. Kopf M, Baumann H, Freer G, Freudenberg M, Lamers M, Kishimoto T, Zinkernagel R, Bluethmann H, Kohler G. Impaired immune and acute-phase responses in interleukin-6-decient mice. Nature 1994: 368: 339 342. 134. Kotake S, Sato K, Kim KJ, Takahashi N, Udagawa N, Nakamura I, Yamaguchi A, Kishimoto T, Suda T, Kashiwazaki S. Interleukin-6 and soluble interleukin-6 receptors in the synovial uids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J Bone Miner Res 1996: 11: 8895. 135. Kotev-Emeth S, Pitaru S, Pri-Chen S, Savion N. Establishment of a rat long-term culture expressing the osteogenic phenotype: dependence on dexamethasone and FGF-2. Connect Tissue Res 2002: 43: 606612. 136. Kouba DJ, Chung KY, Nishiyama T, Vindevoghel L, Kon A, Klement JF, Uitto J, Mauviel A. Nuclear factor-kappa B mediates TNF-alpha inhibitory effect on alpha 2(I) collagen (COL1A2) gene transcription in human dermal broblasts. J Immunol 1999: 162: 42264234. 137. 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: 14721477. 138. Kumar S, Votta BJ, Rieman DJ, Badger AM, Gowen M, Lee JC. IL-1- and TNF-induced bone resorption is mediated by p38 mitogen activated protein kinase. J Cell Physiol 2001: 187: 294303. 139. Kurokouchi K, Kambe F, Yasukawa K, Izumi R, Ishiguro N, Iwata H, Seo H. TNF-alpha increases expression of IL-6 and ICAM-1 genes through activation of NF-kappaB in osteoblast-like ROS17/2.8 cells. J Bone Miner Res 1998: 13: 12901299. 140. Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998: 93: 165176. 141. Lametsch R, Rasmussen JT, Johnsen LB, Purup S, Sejrsen K, Petersen TE, Heegaard CW. Structural characterization of the broblast growth factor-binding protein puried from bovine prepartum mammary gland secretion. J Biol Chem 2000: 275: 1946919474. 142. Laurikkala J, Kassai Y, Pakkasjarvi L, Thesleff I, Itoh N. Identication of a secreted BMP antagonist, ectodin, integrating BMP, FGF, and SHH signals from the tooth enamel knot. Dev Biol 2003: 264: 91105. 143. Lee JK, Kim SH, Lewis EC, Azam T, Reznikov LL, Dinarello CA. Differences in signaling pathways by IL-1beta and IL18. Proc Natl Acad Sci U S A 2004: 101: 88158820. 144. Lerner UH. New molecules in the tumor necrosis factor ligand and receptor superfamilies with importance for physiological and pathological bone resorption. Crit Rev Oral Biol Med 2004: 15: 6481. 145. Lewis DB, Liggitt HD, Effmann EL, Motley ST, Teitelbaum SL, Jepsen KJ, Goldstein SA, Bonadio J, Carpenter J, Perlmutter RM. Osteoporosis induced in mice by overproduction of interleukin 4. Proc Natl Acad Sci U S A 1993: 90: 1161811622. 146. Li YP, Stashenko P. Proinammatory cytokines tumor necrosis factor-alpha and IL-6, but not IL-1, downregulate the osteocalcin gene promoter. J Immunol 1992: 148: 788 794. 147. Littlewood AJ, Aarden LA, Evans DB, Russell RG, Gowen M. Human osteoblast like cells do not respond to interleukin-6. J Bone Miner Res 1991: 6: 141148. 148. Liu D, Xu JK, Figliomeni L, Huang L, Pavlos NJ, Rogers M, Tan A, Price P, Zheng MH. Expression of RANKL and OPG mRNA in periodontal disease: possible involvement in bone destruction. Int J Mol Med 2003: 11: 1721. 149. Lorenzo JA, Sousa SL, Alander C, Raisz LG, Dinarello CA. Comparison of the bone-resorbing activity in the supernatants from phytohemagglutinin-stimulated human peripheral blood mononuclear cells with that of cytokines through the use of an antiserum to interleukin 1. Endocrinology 1987: 121: 11641170. 150. Lynch SE, Williams RC, Polson AM, Howell TH, Reddy MS, Zappa UE, Antoniades HN. A combination of plateletderived and insulin-like growth factors enhances periodontal regeneration. J Clin Periodontol 1989: 16: 545548. 151. Lynch SE, de Castilla GR, Williams RC, Kiritsy CP, Howell TH, Reddy MS, Antoniades HN. The effects of short-term application of a combination of platelet-derived and insulin-like growth factors on periodontal wound healing. J Periodontol 1991: 62: 458467. 152. MacIntyre I, Zaidi M, Alam AS, Datta HK, Moonga BS, Lidbury PS, Hecker M, Vane JR. Osteoclastic inhibition: an action of nitric oxide not mediated by cyclic GMP. Proc Natl Acad Sci U S A 1991: 88: 29362940. 153. MacPherson H, Noble BS, Ralston SH. Expression and functional role of nitric oxide synthase isoforms in human osteoblast-like cells. Bone 1999: 24: 179185. 154. Maeda S, Hayashi M, Komiya S, Imamura T, Miyazono K. Endogenous TGF-beta signaling suppresses maturation of osteoblastic mesenchymal cells. Embo J 2004: 23: 552563. 155. Makiishi-Shimobayashi C, Tsujimura T, Iwasaki T, Yamada N, Sugihara A, Okamura H, Hayashi S, Terada N. Interleukin-18 upregulates osteoprotegerin expression in stromal/osteoblastic cells. Biochem Biophys Res Commun 2001: 281: 361366. 156. Manolagas SC. The role of IL-6 type cytokines and their receptors in bone. Ann N Y Acad Sci 1998: 840: 194204. 157. Mansukhani A, Bellosta P, Sahni M, Basilico C. Signaling by broblast growth factors (FGF) and broblast growth factor receptor 2 (FGFR2)-activating mutations blocks

68

Effects of growth factors and cytokines on osteoblast differentiation


mineralization and induces apoptosis in osteoblasts. J Cell Biol 2000: 149: 12971308. Massague J, Chen YG. Controlling TGF-beta signaling. Genes Dev 2000: 14: 627644. McCulloch CAG, Nemeth E, Lowenberg B, Melcher AH. Paravascuclar cells in endosteal spaces of alveolar bone contribute to periodontal ligament cell populations. Anat Record 1987: 219: 233242. Minuto F, Palermo C, Arvigo M, Barreca AM. The IGF system and bone. J Endocrinol Invest 2005: 28: 810. Miyazono K, Maeda S, Imamura T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev 2005: 16: 251263. Mohammed S, Pack AR, Kardos TB. The effect of transforming growth factor beta one (TGF-beta 1) on wound healing, with or without barrier membranes, in a Class II furcation defect in sheep. J Periodontal Res 1998: 33: 335 344. Mori K, Hatamochi A, Ueki H, Olsen A, Jimenez SA. The transcription of human alpha 1(I) procollagen gene (COL1A1) is suppressed by tumour necrosis factor-alpha through proximal short promoter elements: evidence for suppression mechanisms mediated by two nuclear-factorbinding sites. Biochem J 1996: 319 (Pt 3): 811816. Murakami S, Takayama S, Ikezawa K, Shimabukuro Y, Kitamura M, Nozaki T, Terashima A, Asano T, Okada H. Regeneration of periodontal tissues by basic broblast growth factor. J Periodontal Res 1999: 34: 425430. Murakami S, Takayama S, Kitamura M, Shimabukuro Y, Yanagi K, Ikezawa K, Saho T, Nozaki T, Okada H. Recombinant human basic broblast growth factor (bFGF) stimulates periodontal regeneration in class II furcation defects created in beagle dogs. J Periodontal Res 2003: 38: 97103. Naganawa T, Xiao L, Abogunde E, Sobue T, Kalajzic I, Sabbieti M, Agas D, Hurley MM. In vivo and in vitro comparison of the effects of FGF-2 null and haplo-insufciency on bone formation in mice. Biochem Biophys Res Commun 2006: 339: 490498. Nagy Z, Radeff J, Stern PH. Stimulation of interleukin-6 promoter by parathyroid hormone, tumor necrosis factor alpha, and interleukin-1beta in UMR-106 osteoblastic cells is inhibited by protein kinase C antagonists. J Bone Miner Res 2001: 16: 12201227. Nakahara T, Nakamura T, Kobayashi E, Inoue M, Shigeno K, Tabata Y, Eto K, Shimizu Y. Novel approach to regeneration of periodontal tissues based on in situ tissue engineering: effects of controlled release of basic broblast growth factor from a sandwich membrane. Tissue Eng 2003: 9: 153162. Nakase T, Takaoka K, Masuhara K, Shimizu K, Yoshikawa H, Ochi T. Interleukin-1 beta enhances and tumor necrosis factor-alpha inhibits bone morphogenetic protein-2-induced alkaline phosphatase activity in MC3T3-E1 osteoblastic cells. Bone 1997: 21: 1721. Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, de Crombrugghe B. The novel zinc ngercontaining transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 2002: 108: 1729. 171. Nanes MS. Tumor necrosis factor-alpha: molecular and cellular mechanisms in skeletal pathology. Gene 2003: 321: 115. 172. Nanes MS, McKoy WM, Marx SJ. Inhibitory effects of tumor necrosis factor-alpha and interferon-gamma on deoxyribonucleic acid and collagen synthesis by rat osteosarcoma cells (ROS 17/2.8). Endocrinology 1989: 124: 339345. 173. Nanes MS, Kuno H, Demay MB, Kurian M, Hendy GN, DeLuca HF, Titus L, Rubin J. A single up-stream element confers responsiveness to 1,25-dihydroxyvitamin D3 and tumor necrosis factor-alpha in the rat osteocalcin gene. Endocrinology 1994: 134: 11131120. 174. Nevins M, Camelo M, Nevins ML, Schenk RK, Lynch SE. Periodontal regeneration in humans using recombinant human platelet-derived growth factor-BB (rhPDGF-BB) and allogenic bone. J Periodontol 2003: 74: 12821292. 175. Nevins M, Giannobile WV, McGuire MK, Kao RT, Mellonig JT, Hinrichs JE, McAllister BS, Murphy KS, McClain PK, Nevins ML, Paquette DW, Han TJ, Reddy MS, Lavin PT, Genco RJ, Lynch SE. Platelet-derived growth factor stimulates bone ll and rate of attachment level gain: results of a large multicenter randomized controlled trial. J Periodontol 2005: 76: 22052215. 176. Nohe A, Keating E, Knaus P, Petersen NO. Signal transduction of bone morphogenetic protein receptors. Cell Signal 2004: 16: 291299. 177. OBrien CA, Gubrij I, Lin SC, Saylors RL, Manolagas SC. STAT3 activation in stromal/osteoblastic cells is required for induction of the receptor activator of NF-kappaB ligand and stimulation of osteoclastogenesis by gp130utilizing cytokines or interleukin-1 but not 1,25-dihydroxyvitamin D3 or parathyroid hormone. J Biol Chem 1999: 274: 1930119308. 178. OBrien CA, Lin SC, Bellido T, Manolagas SC. Expression levels of gp130 in bone marrow stromal cells determine the magnitude of osteoclastogenic signals generated by IL-6-type cytokines. J Cell Biochem 2000: 79: 532541. 179. Ohmori Y, Hanazawa S, Amano S, Hirose K, Kumegawa M, Kitano S. Effects of recombinant human interleukin 1 alpha and interleukin 1 beta on cell growth and alkaline phosphatase of the mouse osteoblastic cell line MC3T3E1. Biochim Biophys Acta 1988: 970: 2230. 180. Ohyama Y, Nifuji A, Maeda Y, Amagasa T, Noda M. Spaciotemporal association and bone morphogenetic protein regulation of sclerostin and osterix expression during embryonic osteogenesis. Endocrinology 2004: 145: 4685 4692. 181. Okazaki R, Durham SK, Riggs BL, Conover CA. Transforming growth factor-beta and forskolin increase all classes of insulin-like growth factor-I transcripts in normal human osteoblast-like cells. Biochem Biophys Res Commun 1995: 207: 963970. 182. Oklu R, Hesketh R. The latent transforming growth factor beta binding protein (LTBP) family. Biochem J 2000: 352 (Pt 3): 601610. 183. Okubo K, Kobayashi M, Takiguchi T, Takada T, Ohazama A, Okamatsu Y, Hasegawa K. Participation of endogenous IGF-I and TGF-beta 1 with enamel matrix derivative-stimulated cell growth in human periodontal ligament cells. J Periodontal Res 2003: 38: 19.

158. 159.

160. 161.

162.

163.

164.

165.

166.

167.

168.

169.

170.

69

Hughes et al.
184. Okuda K, Kawase T, Momose M, Murata M, Saito Y, Suzuki H, Wolff LF, Yoshie H. Platelet-rich plasma contains high levels of platelet-derived growth factor and transforming growth factor-beta and modulates the proliferation of periodontally related cells in vitro. J Periodontol 2003: 74: 849857. 185. Ornitz DM, Marie PJ. FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev 2002: 16: 14461465. 186. Osyczka AM, Leboy PS. Bone morphogenetic protein regulation of early osteoblast genes in human marrow stromal cells is mediated by extracellular signal-regulated kinase and phosphatidylinositol 3-kinase signaling. Endocrinology 2005: 146: 34283437. 187. Osyczka AM, Noth U, OConnor J, Caterson EJ, Yoon K, Danielson KG, Tuan RS. Multilineage differentiation of adult human bone marrow progenitor cells transduced with human papilloma virus type 16 E6/E7 genes. Calcif Tissue Int 2002: 71: 447458. 188. Owen M. Uptake of [3H] uridine into precursor pools and RNA in osteogenic cells. J Cell Sci 1967: 2: 3956. 189. Owen M. The origin of bone cells. Int Rev Cytol 1970: 28: 213238. 190. Owen M, Friedenstein AJ. Stromal cells: marrow-derived osteogenic precursors. In: Evered D, Harnett S, editors. Cell and Molecular Biology of Vertebrate Hard Tissues. Chichester: John Wiley & Sons Ltd, 1988: 4253. 191. Palmqvist P, Persson E, Conaway HH, Lerner UH. IL-6, leukemia inhibitory factor, and oncostatin M stimulate bone resorption and regulate the expression of receptor activator of NF-kappa B ligand, osteoprotegerin, and receptor activator of NF-kappa B in mouse calvariae. J Immunol 2002: 169: 33533362. 192. Park YJ, Ku Y, Chung CP, Lee SJ. Controlled release of platelet-derived growth factor from porous poly(L-lactide) membranes for guided tissue regeneration. J Control Release 1998: 51: 201211. 193. Parkar MH, Tonetti M. Gene expression proles of periodontal ligament cells treated with enamel matrix proteins in vitro: analysis using cDNA arrays. J Periodontol 2004: 75: 15391546. 194. Pitsillides AA, Rawlinson SC, Suswillo RF, Bourrin S, Zaman G, Lanyon LE. Mechanical strain-induced NO production by bone cells: a possible role in adaptive bone (re)modeling? FASEB J 1995: 9: 16141622. 195. 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: 143 147. 196. Raines EW, Lane TF, Iruela-Arispe ML, Ross R, Sage EH. The extracellular glycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-AB and -BB and inhibits the binding of PDGF to its receptors. Proc Natl Acad Sci U S A 1992: 89: 12811285. 197. Raisz LG, Martin TJ. Prostaglandins in bone and mineral metabolism. In: Peck WA, editor. Bone and Mineral Research. Amsterdam: Exerpta Medica, 1984: 286310. 198. Ralston SH, Todd D, Helfrich M, Benjamin N, Grabowski PS. Human osteoblast-like cells produce nitric oxide and express inducible nitric oxide synthase. Endocrinology 1994: 135: 330336. 199. 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: 18421853. 200. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature 2005: 434: 843850. 201. Reynolds MA, Aichelmann-Reidy ME. The era of biologics and reparative medicine: a pivotal clinical trial of plateletderived growth factor for periodontal regeneration. J Periodontol 2005: 76: 23302332. 202. Rickard DJ, Gowen M, MacDonald BR. Proliferative responses to estradiol, IL-1 alpha and TGF beta by cells expressing alkaline phosphatase in human osteoblast-like cell cultures. Calcif Tissue Int 1993: 52: 227233. 203. Rifas L, Kenney JS, Marcelli M, Pacici R, Cheng SL, Dawson LL, Avioli LV. Production of interleukin-6 in human osteoblasts and human bone marrow stromal cells: evidence that induction by IL-1 and tumor necrosis factor-alpha is not regulated by ovarian steroids. Endocrinology 1995: 136: 40564067. 204. Ripamonti U, Reddi AH. Tissue engineering, morphogenesis, and regeneration of the periodontal tissues by bone morphogenetic proteins. Crit Rev Oral Biol Med 1997: 8: 154163. 205. Ripamonti U, Heliotis M, Rueger DC, Sampath TK. Induction of cementogenesis by recombinant human osteogenic protein-1 (hop-1/bmp-7) in the baboon (Papio ursinus). Archs Oral Biol 1996: 41: 121126. 206. Robey PG, Young MF, Flanders KC, Roche NS, Kondaiah P, Reddi AH, Termine JD, Sporn MB, Roberts AB. Osteoblasts synthesize and respond to transforming growth factor-type beta (TGF-beta) in vitro. J Cell Biol 1987: 105: 457463. 207. Rodan SB, Wesolowski G, Thomas KA, Yoon K, Rodan GA. Effects of acidic and basic broblast growth factors on osteoblastic cells. Connect Tissue Res 1989: 20: 283 288. 208. Rosen CJ, Dimai HP, Vereault D, Donahue LR, Beamer WG, Farley J, Linkhart S, Linkhart T, Mohan S, Baylink DJ. Circulating and skeletal insulin-like growth factor-I (IGFI) concentrations in two inbred strains of mice with different bone mineral densities. Bone 1997: 21: 217223. 209. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 1999: 4: 611617. 210. Rosenquist JB, Ohlin A, Lerner UH. Cytokine-induced inhibition of bone matrix proteins is not mediated by prostaglandins. Inamm Res 1996: 45: 457463. 211. Rutherford RB, Niekrash CE, Kennedy JE, Charette MF. Platelet-derived and insulin-like growth factors stimulate regeneration of periodontal attachment in monkeys. J Periodontal Res 1992: 27: 285290. 212. Rutherford RB, Ryan ME, Kennedy JE, Tucker MM, Charette MF. Platelet-derived growth factor and dexamethasone combined with a collagen matrix induce regeneration of the periodontium in monkeys. J Clin Periodontol 1993: 20: 537544. 213. Rydziel S, Ladd C, McCarthy TL, Centrella M, Canalis E. Determination and expression of platelet-derived growth factor-AA in bone cell cultures. Endocrinology 1992: 130: 19161922.

70

Effects of growth factors and cytokines on osteoblast differentiation


214. Rydziel S, Shaikh S, Canalis E. Platelet-derived growth factor-AA and -BB (PDGF-AA and -BB) enhance the synthesis of PDGF-AA in bone cell cultures. Endocrinology 1994: 134: 25412546. 215. Rydziel S, Durant D, Canalis E. Platelet-derived growth factor induces collagenase 3 transcription in osteoblasts through the activator protein 1 complex. J Cell Physiol 2000: 184: 326333. 216. Saito M, Yoshida K, Hibi M, Taga T, Kishimoto T. Molecular cloning of a murine IL-6 receptor-associated signal transducer, gp130, and its regulated expression in vivo. J Immunol 1992: 148: 40664071. 217. Saksela O, Moscatelli D, Sommer A, Rifkin DB. Endothelial cell-derived heparan sulfate binds basic broblast growth factor and protects it from proteolytic degradation. J Cell Biol 1988: 107: 743751. 218. Sammartino G, Tia M, Marenzi G, di Lauro AE, DAgostino E, Claudio PP. Use of autologous platelet-rich plasma (PRP) in periodontal defect treatment after extraction of impacted mandibular third molars. J Oral Maxillofac Surg 2005: 63: 766770. 219. Sampath TK, Maliakal JC, Hauschka PV, Jones WK, Sasak H, Tucker RF, White KH, Coughlin JE, Tucker MM, Pang RH, Corbett C, Oppermann H, Rueger DC. Recombinant human osteogenic protein-1 (hOP-1) induces new bone formation in vivo with a specic activity comparable with natural bovine osteogenic protein and stimulates osteoblast proliferation and differentiation in vitro. J Biol Chem 1992: 267: 2035220362. 220. Sanchez AR, Sheridan PJ, Kupp LI. Is platelet-rich plasma the perfect enhancement factor? A current review. Int J Oral Maxillofac Implants 2003: 18: 93103. 221. Schwartz Z, Carnes DL Jr, Pulliam R, Lohmann CH, Sylvia VL, Liu Y, Dean DD, Cochran DL, Boyan BD. Porcine fetal enamel matrix derivative stimulates proliferation but not differentiation of pre-osteoblastic 2T9 cells, inhibits proliferation and stimulates differentiation of osteoblast-like MG63 cells, and increases proliferation and differentiation of normal human osteoblast NHOst cells. J Periodontol 2000: 71: 12871296. 222. Seo BM, Miura M, Gronthos S, Bartold PP, Batouli S, Brahim J, Young M, Robey PG, Wang PC, Shi S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004: 364: 149155. 223. Shimizu E, Nakajima Y, Kato N, Nakayama Y, Saito R, Samoto H, Ogata Y. Regulation of rat bone sialoprotein gene transcription by enamel matrix derivative. J Periodontol 2004: 75: 260267. 224. Sigurdsson TJ, Lee MB, Kubota K, Turek TJ, Wozney JM, Wikesjo UM. Periodontal repair in dogs: recombinant human bone morphogenetic protein-2 signicantly enhances periodontal regeneration. J Periodontol 1995: 66: 131138. 225. Simmons PJ, Torok-Storb B. Identication of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood 1991: 78: 5562. 226. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, DeRose M, Elliott R, Colombero A, Tan HL, Trail G, Sullivan J, Davy E, Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison W, Campbell P, Sander S, Van G, Tarpley J, Derby P, Lee R, Boyle WJ. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997: 89: 309319. Sims JE, Gayle MA, Slack JL, Alderson MR, Bird TA, Giri JG, Colotta F, Re F, Mantovani A, Shanebeck K, Grabstein KH, Dower SK. Interleukin 1 signaling occurs exclusively via the type I receptor. Proc Natl Acad Sci U S A 1993: 90: 61556159. Smit TH, Burger EH. Is BMU-coupling a strain-regulated phenomenon? A nite element analysis. J Bone Miner Res 2000: 15: 301307. Smith DD, Gowen M, Mundy GR. Effects of interferongamma and other cytokines on collagen synthesis in fetal rat bone cultures. Endocrinology 1987: 120: 2494 2499. 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: 20852094. Stashenko P, Dewhirst FE, Rooney ML, Desjardins LA, Heeley JD. Interleukin-1 beta is a potent inhibitor of bone formation in vitro. J Bone Miner Res 1987: 2: 559565. Stashenko P, Dewhirst FE, Rooney ML, Desjardins LA, Heeley JD. Interleukin-1b is a potent inhibitor of bone formation in vitro. J Bone Miner Res 1987: 2: 559565. Suzuki S, Nagano T, Yamakoshi Y, Gomi K, Arai T, Fukae M, Katagiri T, Oida S. Enamel matrix derivative gel stimulates signal transduction of BMP and TGF-beta. J Dent Res 2005: 84: 510514. Taba M, Jr, Jin Q, Sugai JV, Giannobile WV. Current concepts in periodontal bioengineering. Orthod Craniofac Res 2005: 8: 292302. Taguchi Y, Yamamoto M, Yamate T, Lin SC, Mocharla H, DeTogni P, Nakayama N, Boyce BF, Abe E, Manolagas SC. Interleukin-6-type cytokines stimulate mesenchymal progenitor differentiation toward the osteoblastic lineage. Proc Assoc Am Physicians 1998: 110: 559574. Takayama S, Murakami S, Shimabukuro Y, Kitamura M, Okada H. Periodontal regeneration by FGF-2 (bFGF) in primate models. J Dent Res 2001: 80: 20752079. Takayama T, Suzuki N, Narukawa M, Tokunaga T, Otsuka K, Ito K. Enamel matrix derivative stimulates core binding factor alpha1/Runt-related transcription factor-2 expression via activation of Smad1 in C2C12 cells. J Periodontol 2005: 76: 244249. Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J, Wagner EF, Mak TW, Kodama T, Taniguchi T. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell 2002: 3: 889901. Takayanagi H, Sato K, Takaoka A, Taniguchi T. Interplay between interferon and other cytokine systems in bone metabolism. Immunol Rev 2005: 208: 181193. Takiguchi T, Kobayashi M, Nagashima C, Yamaguchi A, Nishihara T, Hasegawa K. Effect of prostaglandin E2 on recombinant human bone morphogenetic protein-2stimulated osteoblastic differentiation in human periodontal ligament cells. J Periodontal Res 1999: 34: 431436.

227.

228.

229.

230.

231.

232.

233.

234.

235.

236.

237.

238.

239.

240.

71

Hughes et al.
241. Tanabe N, Ito-Kato E, Suzuki N, Nakayama A, Ogiso B, Maeno M, Ito K. IL-1alpha affects mineralized nodule formation by rat osteoblasts. Life Sci 2004: 75: 23172327. 242. Tatakis DN, Wikesjo UM, Razi SS, Sigurdsson TJ, Lee MB, Nguyen T, Ongpipattanakul B, Hardwick R. Periodontal repair in dogs: effect of transforming growth factor-beta 1 on alveolar bone and cementum regeneration. J Clin Periodontol 2000: 27: 698704. 243. Taubman MA, Kawai T. Involvement of T-lymphocytes in periodontal disease and in direct and indirect induction of bone resorption. Crit Rev Oral Biol Med 2001: 12: 125135. 244. Teitelbaum SL. Bone resorption by osteoclasts. Science 2000: 289: 15041508. 245. Teitelbaum SL, Ross FP. Genetic regulation of osteoclast development and function. Nat Rev Genet 2003: 4: 638 649. 246. Teng YT, Nguyen H, Gao X, Kong YY, Gorczynski RM, Singh B, Ellen RP, Penninger JM. Functional human T-cell immunity and osteoprotegerin ligand control alveolar bone destruction in periodontal infection. J Clin Invest 2000: 106: R59R67. 247. Thiel S, Dahmen H, Martens A, Muller-Newen G, Schaper F, Heinrich PC, Graeve L. Constitutive internalization and association with adaptor protein-2 of the interleukin-6 signal transducer gp130. FEBS Lett 1998: 441: 231234. 248. Thiel S, Sommer U, Kortylewski M, Haan C, Behrmann I, Heinrich PC, Graeve L. Termination of IL-6-induced STAT activation is independent of receptor internalization but requires de novo protein synthesis. FEBS Lett 2000: 470: 1519. 249. Tokiyasu Y, Takata T, Saygin E, Somerman M. Enamel factors regulate expression of genes associated with cementoblasts. J Periodontol 2000: 71: 18291839. 250. Udagawa N, Horwood NJ, Elliott J, Mackay A, Owens J, Okamura H, Kurimoto M, Chambers TJ, Martin TJ, Gillespie MT. Interleukin-18 (interferon-gamma-inducing factor) is produced by osteoblasts and acts via granulocyte/macrophage colony-stimulating factor and not via interferon-gamma to inhibit osteoclast formation. J Exp Med 1997: 185: 10051012. 251. Ura K, Morimoto I, Watanabe K, Saito K, Yanagihara N, Eto S. Interleukin (IL)-4 and IL-13 inhibit the differentiation of murine osteoblastic MC3T3-E1 cells. Endocr J 2000: 47: 293302. 252. Urist MR. Bone formation by autoinduction. Science 1965: 150: 893899. 253. Urist MR, Strates BS. Bone morphogenetic protein. J Dent Res 1971: 50: 13921406. 254. Viatour P, Merville MP, Bours V, Chariot A. Phosphorylation of NF-kappaB and IkappaB proteins: implications in cancer and inammation. Trends Biochem Sci 2005: 30: 4352. 255. Vidal ON, Sjogren K, Eriksson BI, Ljunggren O, Ohlsson C. Osteoprotegerin mRNA is increased by interleukin-1 alpha in the human osteosarcoma cell line MG-63 and in human osteoblast-like cells. Biochem Biophys Res Commun 1998: 248: 696700. 256. Walsh MC, Choi Y. Biology of the TRANCE axis. Cytokine Growth Factor Rev 2003: 14: 251263. 257. Walsh S, Jefferiss C, Stewart K, Jordan GR, Screen J, Beresford JN. Expression of the developmental markers STRO-1 and alkaline phosphatase in cultures of human marrow stromal cells: regulation by broblast growth factor (FGF)-2 and relationship to the expression of FGF receptors 14. Bone 2000: 27: 185195. Wang EA, Israel DI, Kelly S, Luxenberg DP. Bone morphogenetic protein-2 causes commitment and differentiation in C3H10T1/2 and 3T3 cells. Growth Factors 1993: 9: 5771. Wei S, Kitaura H, Zhou P, Ross FP, Teitelbaum SL. IL-1 mediates TNF-induced osteoclastogenesis. J Clin Invest 2005: 115: 282290. Weintraub H. The MyoD family and myogenesis: redundancy, networks and thresholds. Cell 1993: 75: 12411244. Weitzmann MN, Cenci S, Rifas L, Brown C, Pacici R. Interleukin-7 stimulates osteoclast formation by upregulating the T-cell production of soluble osteoclastogenic cytokines. Blood 2000: 96: 18731878. Westendorf JJ, Kahler RA, Schroeder TM. Wnt signaling in osteoblasts and bone diseases. Gene 2004: 341: 1939. Wikesjo UM, Xiropaidis AV, Thomson RC, Cook AD, Selvig KA, Hardwick WR. Periodontal repair in dogs: rhBMP-2 signicantly enhances bone formation under provisions for guided tissue regeneration. J Clin Periodontol 2003: 30: 705714. Winkler DG, Sutherland MK, Geoghegan JC, Yu C, Hayes T, Skonier JE, Shpektor D, Jonas M, Kovacevich BR, Staehling-Hampton K, Appleby M, Brunkow ME, Latham JA. Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. Embo J 2003: 22: 62676276. Winkler DG, Sutherland MS, Ojala E, Turcott E, Geoghegan JC, Shpektor D, Skonier JE, Yu C, Latham JA. Sclerostin inhibition of Wnt-3a-induced C3H10T1/2 cell differentiation is indirect and mediated by bone morphogenetic proteins. J Biol Chem 2005: 280: 24982502. Wozney JM. The bone morphogenetic protein family and osteogenesis. Molecular Reproduction & Development 1992: 32: 160167. Wozney J. Biology and clinical applications of rhBMP-2. In: Lynch S, Genco R, Marx R, editors. Tissue Engineering: Applications in Maxillofacial Surgery and Periodontics. Chicago: Quintessence, 1999: 103124. Wozney JM, Rosen V, Celeste AJ, Mitsock LM, Whitters MJ, Kriz RW, Hewick RM, Wang EA. Novel regulators of bone formation: molecular clones and activities. Science 1988: 242: 15281534. Yeh LC, Zavala MC, Lee JC. Osteogenic protein-1 and interleukin-6 with its soluble receptor synergistically stimulate rat osteoblastic cell differentiation. J Cell Physiol 2002: 190: 322331. Yoneda S, Itoh D, Kuroda S, Kondo H, Umezawa A, Ohya K, Ohyama T, Kasugai S. The effects of enamel matrix derivative (EMD) on osteoblastic cells in culture and bone regeneration in a rat skull defect. J Periodontal Res 2003: 38: 333342. Yuasa T, Kataoka H, Kinto N, Iwamoto M, EnomotoIwamoto 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: 225232. Zhu Z, Lee CS, Tejeda KM, Giannobile WV. Gene transfer and expression of platelet-derived growth factors modulate periodontal cellular activity. J Dent Res 2001: 80: 892897.

258.

259.

260.

261.

262. 263.

264.

265.

266.

267.

268.

269.

270.

271.

272.

72

Você também pode gostar