<<

2. Osteogenic Growth Factors and Cytokines and Their Role in Repair Louis C. Gerstenfeld, Cory M. Edgar, Sanjeev Kakar, Kimberly A. Jacobsen, and Thomas A. Einhorn

transforming growth factor β (TGF-β) super- 2.1 Introduction family, angiogenic factors, and parathyroid hormone/parathyroid hormone-related peptide Ontogenetic development is initiated at the (PTH/PTHrP). Major emphasis has been time of fertilization and terminates with the directed to these molecules because their activ- differentiation, growth, and maturation of spe- ities constitute current targets of pharmaco- cialized tissues and organs. These developmen- logical studies to promote or alter bone . tal processes are characterized by molecular Short reviews of the fi broblast growth factor specialization that accompanies cellular differ- (FGF) and Wnt families of factors are also pre- entiation and tissue morphogenesis. Most sented in the context of their known functions developmental processes terminate after birth in skeletal development and intended use as or when animals reach sexual maturity, but therapeutic agents. The second half of the some morphogenetic processes are reinitiated review (sections 2.3–5) is focused on the in response to injury in specifi c tissues. One anatomy and cell biology of bone healing, on such regenerative process is the repair of skel- what is known about the temporal and spatial etal fractures and bone tissue after surgery, expression of the various cytokines during a process that recapitulates specifi c aspects bone healing, and how cytokines and morpho- of the initial developmental processes in the gens may therapeutically modify the repair course of healing [58, 209]. Several aspects of process. the postnatal tissue environment of fracture healing, however, are unique and differ from what occurs in embryological and postnatal 2.2 Cytokines, Morphogens, development. Understanding how cytokines and morphogens affect fracture or postsurgical and Growth Factors: healing is essential to the development of The TNF-a Family pharmacological and molecular approaches intended to enhance bone healing after surgery or traumatic injury, as well as to promote skel- 2.2.1 The TNF Family of Cytokines etal tissue engineering. and Their Intracellular Functions The fi rst half of this review (Section 2.2) will focus on several groups of soluble protein TNF was fi rst identifi ed in the early 1980s, and factors that regulate postnatal bone repair: the a large superfamily of related molecules has α (TNF-α) family, the since been identifi ed. So far, 18 members with

17 18 Engineering of Functional Skeletal Tissues

15% to 25% amino acid sequence homology growth, primarily through the activation of the and at least six cell-surface receptors have been nuclear factor κB (NFκB) and c-Jun N-terminal described. The two members of this cytokine kinase (JNK) transcription factors. The dichot- family that have been the most extensively omy of cellular responses to these cytokines characterized are TNF and Fas ligand (FasL). resides in the receptors that are activated and The ligands of this family are all predominantly the downstream signal transduction molecules type II transmembrane proteins. The receptors that interact with these receptors. Signal trans- are all type I transmembrane proteins and are duction is mediated through a two-part system believed to aggregate upon interaction with of docking proteins including MORT/FADD, their ligands. Although the extracellular side TRADD, RIP, and CRADD, which bind to the of the receptors is conserved and composed death domain (DD) of the receptors, and the of cysteine repeats, the cytoplasmic domains of adaptor proteins that have been named TRAFs. the receptors are different and mediate unique Downstream from the coupled responses to activities that lead to a multitude of biological TNFR1 and TNFR2 that mediate cell survival responses through variations in their coupled are the various mitogen-activated protein signal transduction processes. These cytokines (MAP)-related kinases. Downstream from the have been implicated in a wide variety of apoptotic activation of TNFR1 and FAS is the diseases, including tumorigenesis, septic shock, activation of specifi c proteases (caspases) [19, viral replication, , rheumatoid 121, 153, 187]. There is a further bifurcation arthritis, diabetes, and other infl ammatory of the apoptotic cascade, with two separate diseases [19, 121, 153, 187]. Recently, several pathways that can mediate apoptosis: an intrin- therapeutic regimens have been approved that sic (mitochondria-dependent low caspase 8) antagonize TNF-α activity to treat a variety of pathway and an extrinsic (mitochondria- autoimmune diseases, including rheumatoid independent high caspase 8) pathway [185]. To arthritis and Crohn’s disease [163, 184]. Pre- understand the complex regulatory functions liminary studies have also examined whether within a tissue that are mediated through the these approaches can be used to impede the actions of the TNF cytokine family, it is neces- loosening of orthopedic prostheses [37]. sary to defi ne the ligands and to specify the The TNF family members with the most actions of specifi c receptors and the specifi c homogeneity are TNF-α, TNF-β (LT-α), and mechanisms of intracellular transduction LT-β. Both TNF-α ligands and TNF-β (LT-α) within that tissue. are homotrimers, whereas LT-β is a heterotri- mer of (LT-α)1(LT-β)2. There are three receptors in this family: TNFR1/p55/death receptor 1/ 2.2.1.1 TNF Cytokines as Arbitrators of the DR1, TNFR2 (p75), and LT-β receptor. Both Tissue Microenvironment by Selective TNF ligands bind both TNF receptors, but LT- Promotion of Cell Death or Survival β/TNF-α trimers only bind to the LT receptor. FasL is a unique family member and is solely The TNF family of cytokines plays a central recognized by its receptor, FAS/Apo1/DR2 [211]. role in the timing of the immune response, Most cells express TNF-α and its receptors, but namely, when to terminate activation of the the expression of TNF-β and its receptor innate infl ammatory response and initiate the appears to be restricted to T cells and natural acquired immune response, and when to termi- killer cells. TNFR1 (p55) is constitutively nate an innate or acquired response and initi- expressed by almost all cells, but TNFR2 (p75) ate local tissue repair and regeneration. Thus is strongly induced in immune and infl amma- both TNFR1 and Fas mediate activation- tory responses. FasL and Fas are also expressed induced cell death in , T cells, and by many cells but show unique expression B cells [99, 111, 187]. The pathological manifes- during many developmental processes, includ- tations of inappropriate control of the apop- ing the hypertrophy of [72, 174] totic processes in immune function are seen in and the regulation of immune cell differentia- mice that are defi cient in TNFR1, Fas, and FAS/ tion [17, 23, 55, 192]. TNF-α and related cyto- TNFR1. These animals exhibit more severe kines either mediate programmed cell death autoimmune disease and accelerated lym- (apoptosis) or facilitate cell survival and phoproliferation. These responses indicate that Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 19 whereas Fas and TNFR1 receptors both activate 126]. Treatment of human articular chondro- the apoptotic cascade and carry out compensa- cytes with FasL in vitro causes apoptosis. tory or redundant functions, each receptor Because the Fas system is present in growth- mediates a unique set of biological responses plate chondrocytes in vivo, it may play a role in [229]. Thus failure to initiate the programmed apoptosis during endochondral cell death of one or another population of development [6, 83]. In previous studies, carti- immune cells that mediate the transition of the lage cells within the fracture callus [224] have specifi c stages of an immune response leads to been shown to express Fas, and articular chon- a variety of systemic autoimmune pathologies drocytes will undergo programmed cell death [204]. In essence, these cytokines act as the in response to TNF-α [69]. The relationship central arbitrators of a tissue’s microenviron- between the apoptotic process and the normal ment during immune activation. They do so progression of endochondral development can by promoting the survival of one population be observed in pathological conditions such as of cells while causing another to undergo rickets, as well as in the numerous genetically apoptosis. engineered defects that affect growth- The TNF-α family of cytokines has been the development. The hallmark of almost all of primary focus of many immune function these defects is either a foreshortening or an studies, but the death receptor family also plays expansion of the growth plates. Two examples a pivotal regulatory role in many developmen- of factors causing an expansion of the growth tal processes [43]. It is interesting that during plate are vitamin D defi ciency in growing postnatal tissue repair and regeneration these animals and the genetically engineered abla- cytokines directly and indirectly regulate many tion of matrix metalloproteinase 9 (MMP-9) nonimmune cell types downstream from an [210]. Ablation of the PTHrP gene, on the other initial immune response [82]. The signaling hand, causes an osteochondrodysplasia, functions by immune cell cytokines during primarily manifested in an accelerated hyper- postnatal tissue repair derive from functions trophy and removal of the chondrocytes. A carried out during embryogenesis. Alterna- phenomenon common to these very different tively, these cells may initiate postnatal repair pathologies of the endochondral process is that or regenerative processes that replace mecha- in all three the timing or rate of chondrocyte nisms that functioned during embryological apoptosis has been altered. The consequence of development. TNF-α thus functions within an abnormally timed apoptosis is that the skeletal tissues either during the course of microenvironment of the endochondral tissue normal skeletal homeostasis or in response to is altered by retention or loss of the chondro- tissue injury [158]. It does so by acting on both cytes. This is important because osteogenesis, apoptotic and nonapoptotic events within mes- vascular invasion, and marrow formation enchymal cell types found in skeletal tissues. follow in sequence as the chondrogenic cells This includes specifi c types of mesenchymal hypertrophy and undergo apoptosis [120]. precursors [78], osteogenic cells [1], and syno- Thus, in analogy with their role during the vial fi broblasts [64, 137]. immune response, the death receptors and Recent studies have shown that activation of ligands during endochondral development TNF-α and/or NFκB can affect tissue repair, promote the removal of one cell population response to injury, and arthritic pathology by (chondrocytes) and are permissive for osteo- specifi cally inducing the expression of mor- genic and marrow cell populations to move phogenetic factors of the TGF-α family [64]. It into the space previously occupied by the car- may also alter second signal activity of SMADs tilage tissue. that mediate bone morphogenetic protein (BMP) signaling [21, 36, 57]. It is now well established that cartilage cells 2.2.1.2 Role of the TNF-α Family of undergo apoptosis during normal endochon- Cytokines in dral development and during arthritic disease [3, 4, 5, 56, 72]. Currently, three members of the As just discussed, embryologic development TNF family of cytokines have been implicated: and postnatal growth are regulated by ontoge- Fas ligand (FasL), TNF-α, and TRAIL [6, 39, 83, netic and systemic hormonal mechanisms. 20 Engineering of Functional Skeletal Tissues

Fracture and skeletal tissue healing after extreme differences in the avascular microen- surgery, on the other hand, are initiated in vironment of cartilage and bone. Indeed, the response to regulatory mechanisms associated interactions of hematopoietic/lymphopoietic with infl ammation and the innate immune and osteogenic microenvironments in regulat- response [16, 54]. Two discrete types of resorp- ing bone remodeling are emerging as a major tion take place during fracture repair. The fi rst area of research, and changes in cytokines occurs at the end of the endochondral period, that alter lymphopoiesis affect both bone in the course of which mineralized cartilage is homeostasis and immune function [30, 105, removed and primary bone is formed. TNF-α 175, 206]. and its receptors remain largely unexpressed during the initial periods of endochondral dif- ferentiation, but are expressed as the cartilage 2.2.2 The Bone Morphogenetic cells hypertrophy and tissue resorption begins. During this same period, there is an increase Proteins (BMPs) in the concentration of RANKL and osteopro- 2.2.2.1 BMPs and Signaling tegrin (OPG) (two members of the TNF-α superfamily) as well as colony- On the basis of their distinct structural charac- stimulating factor (M-CSF), all key regulatory teristics, BMPs (with the exception of what has factors in osteoclastogenesis [118]. However, been named BMP-1) are members of the trans- other cytokines that are associated with bone forming growth factor β (TGF-β) superfamily. remodeling, including interleukin 1α (IL-1α), This family also includes activins, inhibins, IL-1β, and IL-6 [115], are not expressed. The and growth and differentiation factors (GDFs). other type of resorption occurs during second- BMP-1 belongs to the astacin family of ary bone formation, which follows the endo- metalloendopeptidases and exhibits BMP-like chondral phase. These events are comparable activity by proteolytically activating mixtures to the process of coupled remodeling seen in that contain the proforms of BMP. normal bone homeostasis. During this period, The TGF-β superfamily of pre-proproteins expression of IL-1 and IL-6 increases, whereas displays extensive amino acid sequence homol- the levels of OPG, M-CSF, and RANKL ogy across species and can carry out a wide decline. diversity of biological functions. The proteins These data suggest that the processes medi- share a characteristic pattern of seven con- ating endochondral resorption and the more served cysteine residues within the carboxy- prolonged phase of secondary bone remodeling terminal mature region that are essential for differ and that the resorption of the mineral- the formation of cysteine knot domains. This ized cartilage is more dependent on the activi- tertiary protein structure is thought to be criti- ties of M-CSF, OPG, and RANKL. In contrast, cal for receptor interaction [220]. The mature- bone remodeling appears to depend on the region cysteines are also important in the levels of RANKL and to be coregulated by the formation of intermonomeric disulfi de bonds activities of the cytokines IL-1, IL-6, and TNF- necessary for the formation of physiologically α found in . Differences between functional dimers [119]. As in most secreted bone and cartilage remodeling are apparent proteins, there are numerous potential N- from studies of RANKL (TRANCE)-defi cient linkage glycosylation sites located throughout mice and of mice whose RANKL expression the amino acid sequence. Most BMPs induce was rescued by engineering RANKL expression some level of glycosylation, which varies among in their lymphocytes. When RANKL was species, with mouse BMP inducing the lowest expressed by lymphocytes in the knockout and bovine BMP inducing the highest degree of mice, their osteopetrosis was overcome and glycosylation [183, 212]. development was promoted. However, As an example of a typical BMP structure, it was not possible to correct the chondrodys- BMP-2 is translated as a 396-amino-acid pre- plasia of the epiphyseal and metaphyseal proprotein that contains a 19-amino-acid signal regions. The authors therefore concluded that sequence for targeted secretion, a 263-amino- cartilage and bone possess different mecha- acid proregion, and a 114-amino-acid mature nisms that induce RANKL expression [114]. segment. Within the mature region of BMP-2, In this context it is interesting to note the seven cysteines and one N-linked glycosylation Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 21 recognition site are identifi able. The mature type I receptor phosphorylation in the GS protein has a predicted mass of 14 kDa with an domain by the type II receptor [134]. observed mass of 18 kDa, presumably due to Currently, seven type I receptors, termed glycosylation. The functional protein exists as activin receptor-like kinases (ALKs) 1–7, have a homodimer that is linked by two disulfi de been identifi ed in mammals. ALK-3 (BMP type bridges. There is some speculation on the exis- IA) and ALK-6 (BMP type IB) receptors share tence of heterodimeric complexes in some situ- an 85% amino acid sequence identity in the ations, although in normal physiological kinase domains, and both bind BMP-4, BMP-2, settings homodimeric complexes among the GDF-5, and BMP-7 [149]. Truncated forms of BMPs are most common [183, 217]. Consider- the ALK receptors are currently being used to able amino acid sequence similarity exists examine the role of BMP signaling during the between species for the various family members. development of numerous types of tissues. On Approximately 16 BMPs have been character- the other hand, there are only three BMP type ized, with the majority demonstrating a high II receptors that can interact with BMPs. The percentage of amino acid sequence homology BMPR-II receptor seems to bind exclusively to among the different isotypes, in addition to a BMPs, but the activin types IIA and IIB have high level of amino acid conservation between affi nities for specifi c BMPs (BMP-7, BMP-2 and species [119, 217]. GDF-5), in addition to their activin binding BMPs initiate their signaling at the cell [220]. BMPR-II binds all BMPs weakly by itself, surface through interaction with two distinct with a dramatic increase in the binding serine/threonine kinase receptors: a type I affi nity following recruitment of the type I receptor (50–55 kDa) and a type II receptor receptors. (more than 75 kDa) [220]. It appears that they BMP-2 ligand and receptor interactions have weakly interact with certain members of the been carefully studied (160). During BMP-2 type II receptors independently of type I recep- receptor activation, the BMP-2 protein contains tors, but in the presence of both receptors their two distinct domains that facilitate receptor binding affi nity is increased dramatically [133]. interaction. The fi rst is a large, high-affi nity Following receptor dimerization induced by binding site (termed the “wrist epitope”), which BMP ligand binding, the type II receptor trans- interacts with the BMPR-IA. The second is a phosphorylates the type I receptor, which sub- low-affi nity binding site (termed the “knuckle sequently transmits the BMP signal by epitope”), which interacts with BMPR-II [59]. activation of intracellular Smad (Sma and Mad) The wrist epitopes from monomers (BMPs are proteins. This activation is accomplished by dimeric structures) contribute to the binding of the directed phosphorylation of specifi c serine the BMPR-IA receptor, whereas the knuckle or threonine residues within the Smad pro- epitope from only one monomer binds to teins. The structures of the two receptors are BMPR-IA. The juxtapositioning of these regions similar in that they contain N-glycosylated facilitates a close proximity of the receptors extracellular domains, a single membrane- and initiation of intracellular signaling from spanning domain, and an intracellular serine/ inter-receptor type II phosphorylation to type threonine kinase domain. The extracellular I. Transphosphorylation eventually leads to the domains have several conserved cysteine resi- activation of Smad proteins and signal trans- dues believed to facilitate the formation of mission to target downstream responsive genes essential three-dimensional structures involved [149]. in BMP binding [59]. One distinction between Within the cell BMP signals are transduced the two receptor types is the presence of a by the Smad molecules. To date, eight Smad glycine- and serine-rich domain (GS domain) mammalian proteins have been isolated and found on the type I receptor within the intra- characterized. Smad proteins are the direct cellular N-terminal to the serine/threonine downstream signaling molecules of BMPs and kinase domain. This region is important for the other TGF-β superfamily members and are transmission of the BMP signal to intracellular activated directly by their serine/threonine second-messenger proteins by facilitating the kinase receptors. These proteins can be classi- receptors’ ability to interact with Smad pro- fi ed into three distinct groups based on their teins. This was highlighted in an amino acid intracellular function. The receptor-regulated mutagenesis study linking Smad 7 activation to Smads (R-Smads) are the direct signal 22 Engineering of Functional Skeletal Tissues transducers from the BMP receptor complex or lethality. The complete ablation of BMP-2 following receptor transphosphorylation by homologous recombination resulted in events. Smads 1, 5, and 8 interact with types I embryonic lethality when bred to homozygos- and II BMP receptors and are subsequently ity [228]. These animals had distinct cardiac phosphorylated by the type I receptor within defects consistent with the expression pattern- their COOH-terminus at the conserved SSXS ing of BMP-2 in the extraembryonic mesoderm motif [207]. They are then rapidly released and promyocardium [228]. BMP-2 is expressed from the receptor and subsequently interact in a variety of embryonic nonskeletal epithe- with a common mediator Smad (co-Smad). lial and mesenchymal tissues known to play Smad 4 is the only known co-Smad that signals important roles in morphogenesis [139]. For in both the BMP and the TGF-β transduction example, during limb development high levels pathways [207]. The R-Smad and the co-Smad of transcripts were found in the ventral ecto- proteins form active hetero-oligomeric com- derm and apical ectodermal ridges of the plexes, which can then translocate to the developing limb buds. In addition, BMP-2 nucleus and regulate the transcription of spe- expression was detectable in the developing cifi c downstream genes. The nuclear localiza- heart, whisker follicles (ectodermal placodes), tion of the Smad complexes is dependent on tooth buds (epithelial buds, dental papillae, nuclear localization signals present on Smad 4. and odontoblasts), and craniofacial mesen- Consequently, this protein displays constant chyme [139]. Although other studies have vali- nuclear–cytoplasmic shuttling and is capable dated the importance of BMP-2 during a wide of autonomous nuclear import and export array of mesodermal developmental processes, [218]. The third class of Smad proteins consists the protein also plays important roles in regu- of the inhibitory Smads (I-Smads), Smad 6 and lating the postnatal development of mesenchy- Smad 7, which exert their inhibitory effect by mal skeletal tissues [176]. In animals with a binding to the type I receptor and competing homozygous deletion of the mature coding with the R-Smads for binding to the phosphor- region of the BMP-4 gene, development fails at ylated type I receptor. an extremely early stage. The mice fail to All Smads share two conserved regions develop the necessary primordial germ cells termed Mad homology domains 1 (MH1) and (PGCs) to form a functional embryo [123]. 2 (MH2). MH1 is found in the N-terminal Lawson et al. have shown that BMP-4 pro- portion of the protein, whereas MH2 is in the moter-driven LacZ expression in embryos C-terminal portion, with a linker region of prior to gastrulation results in BMP-4 expres- variable length and amino acid sequence sepa- sion in the extraembryonic ectoderm, followed rating the two domains [150]. The MH2 domain by expression in the extraembryonic meso- contains protein–protein interaction sequences derm [123]. These authors concluded that the and is important in R-Smad/co-Smad oligo- initiation of the germ line in the mouse was merization. The MH1 domain seems to carry dependent on secreted BMP-4 signals from the specifi c DNA-binding sequences necessary to previously segregated, extraembryonic, troph- act at the DNA level in the discrimination of ectoderm lineage. This places BMP-4 function gene regulation. However, a putative “Smad at one of the earliest stages of development consensus sequence” has yet to be determined [123]. However, BMPs do not appear to act [107]. individually but in a coordinated network. For example, the above-mentioned PGC cell gen- eration is directed by more than just BMP-4. 2.2.2.2 BMPs and Developmental In fact, one study has demonstrated that BMP- 2 Regulation is primarily expressed in the endoderm of mouse pregastrula and gastrula embryos and BMPs are considered one of the major groups that the PGC generation in the mouse embryo of morphogenetic factors that mediate pat- is regulated not only by extraembryonic ecto- terning and growth of many tissue types derm-derived BMP-4 and BMP-8B, but also by during embryogenesis and organogenesis. In endoderm-derived BMP-2 [223]. the absence of specifi c BMPs, certain systems BMP-7 has been extensively studied. It is fail to develop, resulting in embryonic defects expressed later during mammalian develop- Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 23 ment, but its function is redundant with that of extraskeletal sites induces de novo formation other BMPs, since knockout animals survive of cartilage and bone [203]. This seminal obser- through gestation. However, BMP-7-defi cient vation led to investigations culminating in the mice die shortly after birth because their extensive purifi cation of the osteoinductive kidneys do not develop normally [86]. In situ activity of demineralized bone matrix (DBM) hybridization analysis has shown that the and the sequencing and cloning of the individ- absence of BMP-7 affects the expression of ual BMPs [35, 166, 217]. The subsequent expres- molecular markers of nephrogenesis, such as sion of BMPs in recombinant systems permitted Pax-2 and Wnt-4, between 12.5 and 14.5 days their use in a variety of animal models, in par- postcoitum [138]. In addition, BMP-7-defi cient ticular to demonstrate their stimulating effects mice have defects in the eye that appear to on the repair of fracture and skeletal defects originate during lens development. Skeletal [53, 67, 221]. Even though exogenous BMPs may patterning defects affect the rib cage, the skull, enhance fracture healing, our understanding and the hind limbs; this shows the wide infl u- of their role in skeletal repair and regeneration ence BMPs have in mammalian development remains incomplete. [138]. The importance of BMPs, however, is Using reverse-transcriptase polymerase restricted neither to skeletal development nor chain reaction (PCR) amplifi cation, Nakase to prenatal development. BMP-2 expression has et al. were the fi rst to demonstrate the temporal been reported to be critical for both extraem- and spatial distribution of BMP-4 in fracture bryonic and embryonic development [101], with healing [157]. In an investigation using a mono- BMP-2 shown to be essential for cranial neural clonal antibody against BMP-2 and BMP-4, crest production. Without it, the skeletal and Bostrom et al. delineated the expression of neural derivatives failed to develop. The im- these BMPs over a 4-week period of fracture portance of BMPs during development has healing [26]. Recently, Cho et al. [38] have been most extensively studied in Xenopus. If shown that specifi c members of the TGF-β BMP-4 signaling is disrupted transgenically by superfamily, including the BMPs, may act in expression of a dominant negative form of its combination to promote the various stages of receptor, the ventral mesoderm is converted to intramembranous and endochondral bone for- a dorsal mesoderm [197]. In situ hybridization mation observed during fracture healing. in Xenopus showed that BMP-4 is expressed in Using ribonuclease protection analysis, this a spatially and temporally restricted manner. study demonstrated that BMP-2 has an early Disruption of the pattern of BMP-4 expression peak in expression on day 1 of fracture healing. by localized microinjections of rhBMP-4 This suggests that BMP-2 may be the most severely disturbed embryonic development upstream mediator in the cascade of BMP [49]. These experiments make it clear that expression. BMP-3 appeared to be preferen- BMP-4 regulates dorsal-ventral patterning in tially associated with intramembranous bone terms of both location and temporal expres- formation, whereas BMP-4, -7, and -8 may sion. As a morphogen, BMP-4 modulates meso- function in recruitment during both dermal patterning by establishing concentration intramembranous and endochondral ossifi ca- gradients that cells detect during migration. tion. Taken together, these studies suggest that Further evidence of the signifi cant role active the coordinated expression of multiple BMPs BMPs play in the control of differentiation and their receptors during fracture healing is comes from experiments that have examined important in both skeletal development and the regulation and responsive expression of skeletal repair. However, the roles of specifi c specifi c BMP antagonists, such as Noggin [44, BMPs during fracture healing need to be 49]. The coordinated expression of BMP antag- investigated. onists interferes with BMP function in somite and limb development [172, 190]. 2.2.3 Angiogenic Factors 2.2.2.3 BMP Function in Skeletal Repair Angiogenesis is the process by which new blood In 1965, Marshall R. Urist demonstrated that vessels are formed from pre-existent vessels. the implantation of demineralized bone at It is important for almost all embryological 24 Engineering of Functional Skeletal Tissues development and in wound healing, because result, the hematopoietic cells are protected the higher metabolic activities of cells within from myelosuppressive stresses [9]. developing and healing tissues increase their nutrient and oxygen requirements [32]. Two classes of angiogenic factors and their 2.2.3.1 The VEGF Family and Receptors receptors are associated with new vessel forma- The VEGF family of genes is currently known tion [60, 130]. These are the vascular endothe- to comprise fi ve related genes: VEGF A, B, C, lial growth factor (VEGF) [61] and the and D and placental growth factor (PlGF). All angiopoietin (Ang) [98] families. of these have some sequence similarity to plate- VEGFs promote vascular permeability and let-derived growth factor (PDGF). The VEGF stimulate mitogenesis in vascular endothelial proteins are roughly 45 kDa in size and exist as cells. In conjunction with the angiopoietins homodimers. Some of the VEGF isotypes bind (see below), VEGF stimulates endothelial-cell to heparin. This enhances retention in the ECM survival by inhibiting endothelial-cell apopto- and presentation to cellular receptors. Of the sis. The VEGFs are produced primarily in genomic subtypes, VEGF A is the most preva- response to hypoxia-induced transcription lent, based on tissue distribution and expres- factors (Hif 1α and Hif 2α), which are expressed sion levels. Selective exon splicing leads to by many stromal and extracellular matrix variants of VEGF A, of which six have been (ECM)-producing cells in tissues with a high identifi ed. They are denoted as VEGF 121, 145, degree of vascularization. Vascular endothelial 165, 183, 189, and 206, based on their amino cells express most receptors for the various acid lengths. Of these, VEGF 121 and 165 appear VEGF isoforms and are the primary responders to be the most commonly expressed, whereas to VEGF. the 165, 189, and 206 variants maintain exons The angiopoietins, like the VEGFs, are that encode the heparin-binding domains expressed by stromal, mesenchymal, and [201]. smooth-muscle cells of larger vessels. Their VEGFs have multiple receptors, including receptors are expressed primarily on endothe- VEGFR1, also known as Flt-1, VEGFR2 (KDR lial cells. Angiopoietins appear to be intimately or Flk1), and VEGFR3 (Flt-4). Each of these involved in vessel remodeling and may play a receptors is characterized by multiple IgG-like particular role in wound-healing and tissue- extracellular domains, and each is coupled to repair situations where there are pre-existent intracellular signaling networks through an vessels [171, 202]. The expression of Ang 2 is intracellular domain that has tyrosine kinase up-regulated by hypoxia and the associated Hif activity. Two other more distantly related mem- 1α factor, VEGF, angiotensin II, leptin, and brane receptors, neuropilin 1 and 2, also inter- estrogen. Ang 2 expression is down-regulated act selectively with various VEGF molecules. by basic fi broblast growth factor (bFGF). TNF- VEGFR1 also exists in a soluble form that lacks α also regulates Ang 2 expression, with up- or the ability for intracellular signaling and down-regulation dependent on the tissue type antagonizes the less soluble form of VEGFR1. [75]. Ang 1 expression, although not extensively Each of the VEGF receptors differs in its inter- characterized as yet, appears to be up- action with the VEGF isotypes. VEGF A inter- regulated in response to hypoxia [167]. acts with VEGFR1 and 2 and both neuropilins, Unlike VEGF, angiopoietins are not mito- VEGF B interacts with VEGFR1 and neuropilin genic but promote cell survival by blocking 1, and VEGF C and D interact with VEGFR2 apoptotic signals. Ang 1 also has strong che- and 3, whereas PlGF only interacts with moattractant properties for endothelial cells VEGFR1. These receptors also have the ability and promotes the adhesion of hematopoietic to signal utilizing a variety of intracellular stem cells. Angiopoietins appear to stimulate pathways and can activate PLC, Ras, Shc, Nck both dissolution and migration of endothelial PKC, and PI3 kinase. cells from pre-existent vessels and, in conjunc- tion with VEGF promote cell survival and sta- 98 171 bilize newly formed vessels in [ , ]. Recent 2.2.3.2 Angiopoietins and Tie Receptors studies have shown Tie 2/angiopoietin signal- ing to regulate the hematopoietic stem-cell qui- Three angiopoietins (Ang 1, 2, and 3/4) have escence niche in the bone marrow niche. As a been identifi ed. They are made up of 498 amino Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 25 acids and have a coil domain that is separated ablation strategies to assess the contribution of by a hinged region from a fi brinogen-like vessel formation to new bone formation. domain. Angiopoietins exist as multiple splic- Administration of a soluble VEGF receptor 1- ing variants and only interact with the Tie 2 immunoadhesin, mFlt(1–3)-IgG, completely receptor. blocked new vessel formation in the growth Angiopoietins bind to Tie 1 and 2 receptors, plates of mice and impaired chondrocyte apop- a tyrosine kinase with immunoglobulin and tosis and trabecular bone formation [66]. epidermal growth factor homology domains. Studies by Gerber et al. (66) have identifi ed Ligand binding induces receptor dimerization, VEGF as the key factor that regulates capillary which causes autophosphorylation of the recep- invasion, growth-plate morphogenesis, and tor, thereby activating its kinase signaling [143]. cartilage remodeling. In mice, systemic inhibi- Other studies have shown that the Tie 1 recep- tion of VEGF during periods of rapid growth tor is proteolytically modifi ed when endothe- has led to inhibition of angiogenesis and to a lial cells interact with VEGF. This suggests decrease in the number of chondroclasts/osteo- some coordination between the signaling clasts/ at the growth plates. Chon- events that are mediated by angiopoietins and droclasts/ belong to the monocyte VEGF. Interestingly, although the Tie receptors cell lineage, express VEGFR, and migrate in are tyrosine kinases, they do not signal through response to VEGFR1-selective ligands. This the MAP kinase system used by VEGF, but indicates that VEGFR1 has a role in monocyte appear to recruit various phosphatases selec- migration [15]. Because osteoblasts express tively, including SHP2, a factor that promotes both VEGF receptors and neuropilin 1 [80], the cell migration by altering activities of focal decrease in osteoblasts at the growth plates in adhesion kinases. anti-VEGF-treated mice refl ects an impairment of VEGFR or neuropilin signaling. This in turn has impaired recruitment and/or differentia- 2.2.3.3 The Role of Angiogenic Factors in tion of these cell types. Thus, VEGF contrib- Bone Development utes importantly not only to angiogenesis, but also to osteogenesis. In mice lacking the VEGF Angiogenesis is important during intramem- gene, the long demonstrate a disturbed branous and endochondral bone formation. vascular pattern at birth, consistent with Vascularization of the growth plate contributes reduced bone growth [140]. Osteoblast and to the coupling of and osteo- hypertrophic chondrocyte development are genesis. Chondrocyte apoptosis and osteoclast also impaired [140]. recruitment and activation are essential termi- VEGFs play an important role in regulating nal stages of cartilage hypertrophy. The osteo- bone remodeling. They do so by attracting clasts resorb the mineralized cartilage and endothelial cells, osteoblasts, and osteoclasts thereby permit bone formation by osteoblasts. [46, 147] and by autocrine regulation of chon- Morphological evidence suggests that chon- drocyte function [33]. Local administration of drocyte apoptosis occurs readily following the VEGF also enhances osteoclast number [100]. invasion of endothelial cells [56, 90] and that Further linkage between VEGF and bone for- chondrocyte death is induced by diffusible mation was recently described by studies in factors that arise either from the vasculature or which hypoxia was shown to drive BMP expres- from hematopoietic elements brought in during sion through VEGF [27]. A number of recent angiogenesis [71, 72]. The newly developing studies have also shown that BMPs stimulate blood vessels in addition establish the conduit the expression of VEGF by osteoblasts and for the cells that form primary bone following osteoblast-like cells [47, 222]. Finally, the tissue- resorption of the mineralized trabeculae of specifi c regulation of VEGF expression during cartilage [59]. bone development seems to be dependent on The interrelationship between blood-vessel the expression of Cbfa1/Runx2, known as the formation and osteogenesis has been studied key transcriptional factor that regulates the by various approaches aimed at inhibiting commitment of mesenchymal cells to the skel- VEGF signaling. Because mice whose VEGF etal-cell lineage [226]. Taken together, these has been ablated die as embryos, studies have fi ndings provide a considerable body of evi- utilized inhibitors of VEGF signaling or select dence in support of the concept that VEGF 26 Engineering of Functional Skeletal Tissues mediates bone formation by direct stimulation healing studies, showed that during fracture of osteogenesis and indirectly by its effects on healing Ang 2 was the factor with the highest vascularization. expression. Unlike the VEGF family, which promotes new vessel formation by stimulating 2.2.3.4 The Role of Angiogenic Factors in endothelial cell division, Ang 2 promotes desta- Tissue Healing bilization and regression of blood vessels in the absence of VEGF A or bFGF [87, 135, 141]. Fracture healing and bone or tissue repair Recent fi ndings have suggested that Ang 2, result in an up-regulation of blood fl ow, so that along with VEGF, promotes new vessel forma- bone regeneration can occur within the callus tion by inducing remodeling of the capillary or repair tissues [10, 52, 179]. The importance basal lamina and by stimulating endothelial- of vascularization during fracture repair was cell sprouting and migration [141]. This sug- confi rmed by studies showing that broad- gests that Ang 2 expression plays a role similar spectrum angiogenic inhibitors completely to that of VEGF in bone repair. By itself, Ang 2 prevented fracture healing, callus formation, inhibits blood-vessel formation, but in combi- and the formation of periosteal woven bone nation with VEGF it stimulates new vessel for- [84, 195]. In contrast, treatment of healing frac- mation and plasticity in existing vessels. tures with VEGF improved bone healing and These studies also pointed to collaborative led to more rapid mineralization of the callus interactions between VEGI (vascular endothe- and regaining of mechanical strength [195]. lial growth inhibitor)-induced angiogenesis The role angiogenesis plays in osteogenesis and the TNF-α family of regulators. Interac- following distraction rupture has been exten- tion of VEGI with death receptor 4 and the sively studied with the aid of an artifi cially pro- primary regulator of the progression of vascu- duced gap following osteotomy. When this larization refl ects a dual role: maintaining technique is used, new bone forms primarily growth arrest of endothelial cells in G0/G1 via an intramembranous mechanism with interfaces, while at the same time inducing extensive revascularization of the regenerated apoptosis in cells that enter the S phase [79, 225, bone. Within the marrow space, venous sinu- 227]. Taken together, these results suggest that, soids are formed that parallel the newly grown after injury, vessels are dissociated into a pool trabeculae. Analysis of experimental models of of nondividing endothelial cells through the osteogenesis following distraction has revealed actions of Ang 2. They are then are held in this an early intense vascular response, with the state through the actions of VEGI, which stim- newly formed vessels maturing into sturdier ulates apoptosis of all cells that enter the S vessels capable of withstanding the tensile phase. When endochondral remodeling is forces that are generated in the distraction gap initiated, VEGF levels rise, stimulating cell [129, 179]. division and allowing endothelial cells to con- As discussed above, angiogenesis appears to tribute to neoangiogenic processes. The concept involve two separate pathways: a VEGF-depen- of controlled cell regression and growth is also dent pathway and an angiopoietin-dependent consistent with the role that angiopoietin is pathway. Interestingly, both Ang 1 and Ang 2 thought to play in blood-vessel formation [87]. have been identifi ed in bone cells during devel- opment [89] and in bone cells that arise in osteogenesis following distraction and fracture 2.2.4 Parathyroid Hormone (PTH)/ healing [34, 127, 128]. Indeed, in studies of mice that had undergone distraction fracture, Ang 1, Parathyroid Hormone-Related Ang 2, and their Tie receptors were expressed Peptide (PTHrP) and throughout healing at the same time that VEGF A and VEGF C were expressed. These regula- PTHrP Signaling tors of angiogenesis were expressed throughout 2.2.4.1 PTH Versus PTHrP: Endocrine Versus the chondrogenic phase of healing, reaching Paracrine Effects maximum levels during the late phases of endochondral remodeling and during bone PTH, a peptide, is an hormone that is synthe- formation. These studies, as well as fracture- sized by the parathyroid gland. The mature Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 27 form of the peptide is 84 amino acids in length. 2.2.4.2 PTH Receptor Signal Transduction PTHrP, on the other hand, is an autocrine/ and Nuclear Effects paracrine factor that was fi rst discovered as the primary cause of malignant hypocalcemia in Receptor activities are modulated through many cancers [28]. It is normally expressed interaction with heterodimeric (α, β, γ) G pro- during development and in many postnatal teins that activate or inhibit cyclase production tissues, including cardiac, vascular, mammary, of cAMP. The levels of cAMP then control the cartilage, and renal tissues, as well as a number activity of protein kinase A (PKA), which serves of other epithelial surfaces. The mature form of as the cAMP intracellular second signal trans- PTHrP is 141 amino acids in length. Even ducer [77]. The activation of the receptor by though PTH and PTHrP bind to the same ligand binding also activates phospholipases receptor, the two molecules share only a limited Cβ through Gαq11. Activated phospholipase sequence homology along the fi rst 34 amino generates diacylglycerol and 1,4,5-inositol tri- acids of their amino terminal sequences phosphate (IP3). These two molecules activate and diverge considerably in their carboxyl both protein kinase C (PKC) and Ca2+ release. domains. Study of the “cross-talk” between the PKA- The effects of the major calcitropic hormone arm, PKC kinases [125], and Ca2+ will likely sort PTH on skeletal cells are very important clini- out the many parallel and sometimes antago- cally, owing to the role played by the skeleton nistic functions of the PTH and PTHrP ligands in mineral homeostasis. Both molecules have in different target-cell populations [76]. similar systemic effects on mineral metabo- At the nuclear level, both the PKA and the lism, yet they differ in amino acid composition PKC families of kinases mediate their actions and physiological function. PTH and PTHrP through the phosphorylation of members of share a common receptor (PTHR1) and, when the leucine zipper family of transcription in the circulation, are primarily targeted to the factors [125]. These transcription factors, when kidney and skeleton [62]. In the kidney, PTH phosphorylated, may activate or inhibit the and PTHrP bring about their calcitropic effects transcription of specifi c genes [113, 132] and by stimulating calcium reabsorption and phos- may be classifi ed into two broad groups: the phate excretion in the distal end of the collect- cAMP response element-binding protein family ing tubules. They also regulate formation of (CREBs) and members of the AP-1 family. The the active vitamin D3 metabolite, 1,25- CREBs include the CREB, CREM, and ATF dihydroxyvitamin D3 by activating the enzyme classes of factors; the primary members of the that carries out the 1α-hydroxylation of 25- AP-1 family include fos, jun, and fra [8, 77, 125, hydroxyvitamin D3 in the proximal tubules. 131, 173, 205]. In general, the actions of PKA are This leads to a rise in serum calcium and a mediated through the phosphorylation of lowering of phosphate level. The effects on the members of the CREB family, while PKC skeletal system are less well understood. PTH appears to act on members of the AP-1 family. binds to the receptors of osteoblasts [177], However, phosphorylation may not be restricted which produce paracrine factors that induce to one type of kinase or individual factors. The increased activation and recruitment of factors are active when dimeric. Members of osteoclasts. both families can undergo specifi c heterodi- PTH and PTHrP, like other peptide hor- merization with one another [125]. Heterodi- mones, mediate their effects through interac- merization gives rise to a diversity of specifi c tion with a receptor. Two forms of this receptor transcription factors. As a result, genes may are known, but the two peptides interact pri- be expressed or silenced in a tissue-specifi c marily with PTH1R. This receptor has seven fashion in response to common second signals transmembrane domains and is closely related [77]. Similarly targeted changes or ablation of to a subset of similar receptors that include the these transcriptional regulators give rise to calcitonin and secretin receptors [65]. PTH and specifi c skeletal tissue phenotypes. PTHrP bind almost identically to the receptor, Extensive data have been accumulated to which has both endocrine and autocrine/para- suggest that the leucine zipper family of tran- crine functions in the tissues in which it is scription factors plays a major role in the regu- expressed [2]. lation of gene expression and development in 28 Engineering of Functional Skeletal Tissues the skeleton. Studies in which both the c-fos PTHrP in their growth plates, which are made and the v-fos genes were virally introduced up mainly of hypertrophic chondrocytes. Con- have shown that fos expression generated osteo- versely, animals lacking PTHrP have very small sarcomas [74, 180, 181]. C-fos knockout mice zones of proliferating chondrocytes and exhibit develop osteochondrodysplasia, overproduce a premature transition to cellular hypertrophy hypertrophic cartilage, and cannot replace and mineralization [136, 194]. Interestingly, if bone. This condition in some ways looks like the Ihh-ablated mice are engineered to have a osteopetrosis [213]. Other studies have also constitutively active PTH1R receptor, prema- shown increases in c-fos proto-oncogene in ture chondrocyte hypertrophy is prevented, bone from patients with fi brous dysplasia in but proliferation of the chondrocytes in the whom bone formation is overexpressed or bone growth zones is still diminished. These fi nd- forms ectopically [31]. Studies examining dif- ings suggest molecular mechanisms that are ferent members of the basic leucine zipper regulated by Ihh and not activated by PTHrP protein family have demonstrated that both control cell proliferation [103]. ATF-2 and hXBP are expressed in skeletal A further understanding of the developmen- tissues [40, 173]. Ablation of ATF-2 leads to a tal role of PTHrP has been gained from studies defect in endochondral ossifi cation with a his- in two different human chondrodysplasias. topathology similar to human hypochondro- One type of chondrodysplasia is a nonlethal, plasia [173]. autosomal dominant disorder that was fi rst identifi ed by Jansen in 1934 [95]. It has subse- 2.2.4.3 The Role of PTHrP in quently been characterized at a molecular level Endochondral Development as a constitutively activating mutation in the PTH1R receptor [186]. The patients are charac- The discovery of PTHrP as the primary factor terized by short limbs caused by severe abnor- in malignant hypocalcemia constituted a malities in their growth plates and associated major advance in understanding the systemic hypocalcemia. The second type of chondrodys- effects of many malignancies. However, the plasia was identifi ed by Blomstrand et al. [22]. subsequent characterization of PTHrP as an It is a prenatal lethal chondrodysplasia charac- essential autocrine/paracrine factor in skeletal- terized by abnormal bone ossifi cation and tissue development was equally important. shortened limbs. In these fetuses, the limbs Initial animal studies demonstrated that PTHrP show very advanced endochondral develop- mRNA was fully expressed in perichondral ment, and the disease is characterized by an cells and in the chondrocytes found in the autosomal recessive pattern of inheritance. proliferating zones of endochondral growth Molecular analysis of these patients suggested plates. The PTHrP receptor was expressed the disease is due to an inactivating mutation progressively more fully as endochondral in the PTH1R receptor [97]. chondrocytes matured toward their terminal hypertrophic state. Missense expression studies of PTHrP in developing avian embryo growth 2.2.4.4 PTH as a Therapeutic for plates and studies in transgenic animals with Osteoporosis and Augmentation the targeted ablation of PTHrP have demon- of Fracture Healing strated a complex negative feedback loop that involves Indian hedgehog (Ihh) regulation of Numerous recent studies have focused on the the progression of chondrocyte development systemic effects and potential therapeutic appli- during endochondral bone formation [102, 122, cations of PTH [48, 159, 169]. The continuous 208]. These studies demonstrate that Ihh posi- infusion of PTH into mammals induces cata- tively regulates PTHrP expression, as a result bolic events, increases bone remodeling, and of which cells are maintained in an undifferen- leads to a loss of skeletal bone mass. On the tiated state, with Ihh promoting proliferation other hand, intermittent dosing seems to have and thereby expanding the population. The anabolic effects and results in increased bone increased output of PTHrP causes Ihh expres- mass [116, 151, 199]. Clinical trials utilizing the sion to be down-regulated by expanding the 1–34 PTH peptide have increased bone mineral pool of proliferating immature chondrocytes. density and reduced the risk of vertebral and Ihh-ablated transgenic mice have no detectable nonvertebral fractures in postmenopausal Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 29 women. Intermittent treatment also has im- current family of closely structurally related proved the bone mass in osteoporotic men [85]. proteins is encoded by at least 22 genes. Four The recent approval of PTH(1–34) as an ana- distinct FGF receptors each a unique gene bolic treatment for osteoporosis has been a product, mediate activity through tyrosine major impetus to the use of PTH in bone kinase activity. Each receptor appears to be healing. PTH administration has enhanced activated by all members of the FGF ligand early fracture healing in parathyroidectomized family. The ligands in general have heparin- rats [63], with PTH doses ranging from 10 to binding activity and, when complexed with 200µg/kg having signifi cantly improved the heparin, have improved activity. FGF ligands mechanical and histological aspects of normal regulate a wide variety of cellular functions fracture repair in the rat [7, 88, 155]. PTH and can act as mitogens, chemoattractants, analogs have also been shown to reverse the and mediators of cellular differentiation. FGF inhibition of bone healing in ovariectomized receptor activity appears to directly regulate rats [112] and in corticosteroid-treated rabbits the expression of a number of different pro- [25]. PTH(1–34) is reported to increase bone teins, including metalloproteinases and ingrowth and pullout strength in porous metal- morphogens [142, 164, 165]. lic implants [193]. The roles played by FGF in skeletal develop- One drawback of the rat studies is that the ment have been elucidated by identifying auto- hormone doses were much higher than would somal dominant mutations that constitutively be tolerated in humans. To evaluate the clinical activate the FGF receptors [191, 219]. Mutations potential of PTH for fracture healing, patient- in the receptors lead two types of disorders. appropriate doses of recombinant human para- One, in FGFR3, affects axial long-bone thyroid hormone [PTH(1–34); teriparatide; development and leads to the dwarfi ng chon- ForteoTM] were used in a well-established rat drodysplasia syndromes. These include hypo- model. As early as day 21 of this study, calluses chondroplasia [18], achondroplasia [191], and from the group treated with 30µg/kg of PTH thanatophoric dysplasia [178]. The second showed signifi cant increases over controls in group of mutations, in FGFR2, causes a variety terms of torsional strength, stiffness, bone of craniosynostosis syndromes, including the mineral content (BMC), bone mineral density Apert syndrome [215] and the Crouzon syn- (BMD) and cartilage volume. By day 35, both drome [93]. To date, changes in growth due the 5-µg and the 30-µg/kg PTH-treated groups to inactivating mutations in individual FGF showed signifi cant increases in BMC, BMD, and ligands have not been identifi ed. This suggests total osseous tissue volume; the experimental that the developmental functions of the FGF groups also showed signifi cant decreases in ligands involve collaboration among various void space and cartilage volume. At day 35, tor- molecules. sional strength was also signifi cantly increased FGF family signaling pathways play multiple in the group treated with 30µg of PTH. Even and essential roles in the early stages of skeletal after 84 days, the group that had received 30µg patterning and in the recruitment and ultimate of PTH for 21 days, with treatment discontin- apoptosis of mesenchymal cells. They also ued thereafter, exhibited increases in torsional seem to participate in the control of endochon- strength and BMD over comparable control dral growth in the axial skeleton and of cranial values. Thus, daily systemic administration of bone growth at suture lines. During early limb- a low dose of PTH(1–34) enhanced fracture bud development, FGF signaling plays a role in healing and induced an anabolic effect through- mesenchymal epithelia [144]. As a result, FGF- out the entire remodeling phase. 10 is produced and acts on the FGF receptor 2b in the apical ectodermal ridge. Cells in the latter then express FGF-8, which signals back 2.2.5 Other Growth Factors Within to FGFR1c in the limb mesoderm. The role of FGF signaling in endochondral Skeletal Tissues growth has been made apparent by activating 3 2.2.5.1 Growth Factor (FGF) mutations in FGFR . However, the exact effect of the signaling pathways involved in endo- FGFs were originally isolated as oncogenes and chondral development and the downstream shown to stimulate cell proliferation [196]. The FGF signaling on chondrocytes and osteoblasts 30 Engineering of Functional Skeletal Tissues is less well understood. As with BMPs, multiple dogs, a single dose of bFGF injected into the forms of the FGFs are expressed in the peri- fracture sites resulted in increased callus area chondrium. FGFR1 is expressed in prehyper- and BMC and signifi cant recovery in strength trophic and hypertrophic zones and FGFR3 by by week 16. Thus, FGF has therapeutic poten- proliferating chondrocytes. More recently, the tial to enhance bone healing after surgery or actions of FGF signaling have been shown to injury. depend on the stage of chondrocyte differentia- tion and the nature of the individual ligands. 2.2.5.2 Wnts (Wingless) Thus, specifi c receptors are expressed and interact with specifi c ligands in chondrocytes Wnts are 39- to 46-kDa cysteine-rich, secreted only at specifi c stages of differentiation. Studies glycoproteins that are closely associated with of limb cell cultures have indicated that FGF both the cell surface and the ECM [161, 214]. signaling interacts with both the Ihh/PTHrP Wnts are considered one of the major morpho- and the BMP signaling systems in a complex genetic gene families responsible for appropri- network. FGF signaling seems to accelerate ate embryonic development [146]. Genetic both the onset and the pace of hypertrophic studies fi rst performed in Drosophila have differentiation, in actions that are antagonistic defi ned the function of this gene family. In Dro- to those of BMPs, and to regulate chondrocyte sophila, the wingless gene is required for Ihh expression and hypertrophic differentia- normal patterning in the adult and larval body tion. BMP, on the other hand, seems to rescue segments [13, 14, 162]. The lack of this wingless the remaining proliferating and hypertrophic gene results in the deletion of the posterior chondrocytes in achondroplastic mice. This region of each body segment [14, 162]. Ectopic has led to the conclusion that the interaction of gene expression in Xenopus and gene knockout BMP and FGF in the growth cartilage regulates models in mice have since led to further under- the rate of chondrocyte differentiation and standing of the crucial role that Wnts play in proliferation [148]. organ development, segmentation, CNS pat- The intracellular effects of FGFs are medi- terning, cell fate and growth, limb develop- ated by two signaling pathways: the mitogen- ment, and organization of asymmetric cell activated protein kinase/ERK kinase 1 (MEK1) divisions [11, 45, 168, 216]. To date, approxi- pathway and the Janus kinase-signal trans- mately 100 Wnt genes have been identifi ed in ducer and activator of transcription (JAK- species ranging from Caenorhabditis elegans to STAT) pathway [152, 165, 182]. The JAK-STAT humans [216]. signaling pathway mediates the ability of FGF Once the Wnt proteins are secreted, they signaling to inhibit chondrocyte proliferation bind to two families of cell-surface receptors, and enhances hypertrophic chondrocyte apop- the Frizzled (Fzd) receptors and the low-density tosis, whereas the MEK1 pathway mediates FGF lipoprotein (LDL) receptor-related proteins inhibition of hypertrophic differentiation. (LRPs). The Fzd receptor generally consists of A number of studies have examined whether an extracellular cysteine-rich domain (CRD) FGF has utility in promoting bone formation. that binds the specifi c Wnt protein. This recep- Systemic low doses of basic FGF (FGF-2) stim- tor also consists of a seven membrane spanning ulate endosteal and endochondral bone forma- domain on the cytoplasmic tail towards the tion, but depress periosteal bone formation in carboxy-terminus of the protein. In contrast, growing rats [145, 156]. Local administration the LRP-5 and -6 receptors have a single trans- of acidic FGF (FGF-1) increases new bone for- membrane domain [200]. A variety of secreted mation and , whereas systemic proteins, such as Frizzled-related proteins FGF-1 appears to restore bone microarchitec- (sFRPs), Wnt inhibitory factor 1 (WIF1), and ture and prevent bone loss associated with Cereberus, have been shown to be moderators estrogen withdrawal [50]. Both FGF-1 and of extracellular Wnt signaling. The Dickkopf FGF-2 appear immediately at injury sites after (DKK) protein also exerts regulatory action by fracture. FGF-2 was shown to improve bone directly binding to the LRPs, thereby blocking healing in a study that induced a large seg- signal transduction [11]. mental defect and in another with a metaphy- When the ligand becomes bound to the Fzd seal fracture. In a 32-week study of beagle receptor, three signaling pathways are acti- Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 31 vated: the Wnt/β-catenin (canonical) pathway, 110]. When Wnt 3a expression was analyzed in the Wnt/Ca2+ pathway, and the Wnt/polarity a murine knockout model, severe skeletal pathway. The latter two are defi ned as nonca- defects were observed [91, 198]. Studies of the nonical [11]. It is of interest that a given Wnt direct effects of Wnt 3 on mesenchymal stem protein can activate more than one signaling cells (MSCs) demonstrated that exogenous cascade. The canonical pathway involves stabi- addition of Wnt 3 to murine MSCs inhibited lization of β-catenin, followed by translocation osteogenic differentiation and decreased matrix to the nucleus where transcription genes are mineralization; however, the suppression of activated via the TCF/LEF1 family of transcrip- osteogenesis can be fully reversed when Wnt 3a tion factors [24]. is removed. The noncanonical signaling is not as well The noncanonical effects of Wnt signaling understood, though studies in Drosophila and have been examined through studies of Wnt 5. C. elegans are being continued. The Wnt/Ca2+ In contrast to the inhibitory effects of Wnt 3a, pathway is thought to induce an increase in Wnt 5 appeared to promote osteogenic differ- intracellular Ca2+ and activation of PKC, but entiation of the MSCs. These fi ndings suggest further signaling steps have not yet been iden- that canonical Wnt signaling functions to tifi ed. Genetic studies in Drosophila indicate maintain an undifferentiated, proliferative that the c-Jun N-terminal kinase (JNK) pathway MSC population, whereas the noncanonical is involved in the Wnt/polarity pathway, which Wnts stimulate osteogenic differentiation [24]. in turn regulates cell polarity by controlling Interestingly, ectopic expression of Wnt 5a cytoskeletal organization, utilizing at some delayed chondrocyte maturation and collagen stage the disheveled (Dsh) scaffold protein [11]. type X expression, processes involved in carti- The exact mechanism by which the LRP-5 and lage formation [81, 109]. -6 coreceptors function is not understood, but Many questions remain on the functional they are essential for appropriate signaling. role of the Wnts, their receptors, their intracel- Loss of function of Arrow, the Drosophila lular signaling, and their possible interaction analog to the vertebrate LRP receptor, mimics with other morphogenic factors, such as the the wingless mutation that was fi rst observed TGF-β family. in the early 1980s and therefore provides evidence for the synergism between these receptors [200]. The roles Wnt signaling plays during skeletal development and postnatal bone repair were 2.3 Origins of Postnatal recognized as a result of mutations in humans. One is the autosomal recessive disorder osteo- Skeletal Stem Cells, porosis pseudoglioma, characterized by low Cytokines, and bone mass, frequent deformations and frac- tures, and defects in eye vascularization, all of Morphogenetic Signals which are linked to mutations in LRP-5 [73]. Children with osteoporosis pseudoglioma During Bone Repair have normal endochondral growth and bone turnover, but their trabecular bone volume Bone is unique in that after fracture or surgery, is signifi cantly decreased [106]. Furthermore, it can regenerate the original structure and bio- gain-of-function experiments in humans and mechanical competency of the damaged tissue. in mouse models have shown that organisms Bone repair involves four stages that overlap with an activated LRP-5 mutation exhibit a and cause the various tissue types to interact, high bone mass [12]. as shown in Fig. 2.1. The fracture line in the Because the canonical signal transduction bone determines the spatial relationships of the pathway is fairly well known, Wnt 3a was morphogenetic fi elds during tissue regenera- studied in transgenic mice. Previous studies tion. This is evidenced by the development of had shown that Wnt 3a acts in the apical ecto- two circular centers of cartilage (ECB) that dermal ridge of the limb bud to keep cells in an form symmetrically with respect to the fracture undifferentiated and proliferative state [108, line and taper proximally and distally along the 32 Engineering of Functional Skeletal Tissues

ORIGIN OF CELLS STAGES OF FRACTURE REPAIR & SIGNALS Biological Processes

Muscle D. Initial Injury

Inflammation Marrow response

Marrow Hematoma MSC recruitment

Cortical Bone

E. A. Endochondral Formation Cartilage formation

Periosteal Response ECB Vascular ingrowth Intramembranous bone Formation

F. IMB Primary Bone Formation Bone cell recruitment Chondrocyte apoptosis B. Matrix proteolysis Osteoclast* recruitment Endochondral neovascularization

G. Secondary Bone Secondary Bone Formation Establishment of marrow Osteoclast remodeling Coupled osteoblast recruitment

C. Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 33 bone cortices (see the middle microphotograph MSCs may also originate in the surrounding in Fig. 2.1A, B, and E). At the same time, a muscle or marrow space. Data to support a crescent-shaped region of intramembranous muscle origin come from studies showing that bone formation appears at the proximal and demineralized bone powder or purifi ed BMPs, distal ends of the area of periosteal response when implanted or injected into muscle tissue, and tapers inward toward the fracture line deep induce bone formation [92, 96, 203]. Other in the cartilage ring. Thus, endochondral and studies have shown that a variety of premyo- intramembranous bone formation both con- genic cell lines can differentiate into chondro- tribute to bone healing. genic or osteogenic cells when treated with During bone repair, cell interactions are ini- BMPs [41, 70, 104]. Marrow stroma also can tiated between the external soft tissues that differentiate into osteoblasts and chondrocytes surround the injured bone, the underlying [20, 94, 188, 189]. Once recruited, their numbers cortical bone and marrow, and the developing increase as a result of other morphogenetic endochondral and intramembranous bone factors. It is important to identify the source of tissues (Fig. 2.1A). The origin of the MSCs that the stem cells, because they make up much of contribute to bone repair and the identity of the the callus tissue and may make up as much as cells that initiate morphogenetic signals are 30% of the original volume of the uninjured still unresolved. Figure 2.1 shows potential long bone sources of cells and signals that lead to the con- Vascular tissues grow into the developing struction of these developmental fi elds. callus as new periosteal bone develops and pro- MSCs involved in fracture repair may origi- gresses toward the fracture line from the proxi- nate in the periosteum, the surrounding tissues, mal and distal edges. The interaction of the or both (Fig. 2.1A). The periosteum appears to vascular elements and the initiation and propa- be the primary source of MSCs that then give gation of the periosteal response thus appear rise to the intramembranous bone that forms to be the primary driving mechanisms that in the callus [154]. If the periosteum is removed, facilitate intramembranous bone formation. callus development is diminished [29], because Perivascular mesenchymal cells in blood-vessel periosteal cells robustly produce BMPs during walls may also contribute to this process [27]. the initial phases of fracture healing [26]. These Figure 2.2 summarizes the mesenchymal observations suggest that morphogens recruit lineage and types of morphogens that are stem cells locally and induce them to involved in lineage selection, expansion, sur- differentiate. vival, and programmed cell removal.

Figure 2.1. Anatomic characterization of fracture repair. Left panels (A-C) show an overview of the morphogenetic fields of tissue development and the proximate tissue interactions. (A) Histological section of the fracture site immediately postfracture. Potential tissue origins of mesenchymal stem cells (MSCs) and morphogenetic signals are denoted by the arrows and denoted in the figure. (B) Histological section of the fracture site at 7 days postfracture. The two types of bone-formation processes are denoted as endochondral bone (ECB) and intramembraneous bone (IMB) formation. The two proceed in a symmetrical manner around the fracture site. (C) Histological section of the fracture site at 28 days postfracture. Secondary bone formation and coupled remodeling predominate in the late stage of bone repair. Right panels (D-G) show a summary of the multiple stages of fracture healing. Histological sections are presented for each stage, and the various processes associated with each stage are summarized. All histological specimens are from sagittal sections of mouse tibia transverse fractures and were stained with safranin O and fast green; micrographic images are at 200× magnification. (D) Section for the initial injury was taken from the fracture site 24 hours postinjury. (E) Section depicting the initial periosteal response and endochondral formation is from 7 days postinjury. Arrows denote vascular ingrowth from the peripheral areas of the periosteum. (F) Section depicting the period of primary bone formation is from 14 days postinjury. Arrows denote neovascular growth areas in the underlying new bone. Inset depicts images of an osteoclast (*chondroclast) resorbing an area of calcified cartilage. (G) Sections depicting the period of secondary bone formation are from 21 days postinjury. Callus sites. Inset depicts 400× images of an osteoclast resorbing an area of primary bone. Reproduced with permission from Gerstenfeld LC, Cullinane DM, Barnes GL, et al. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem. 2003 Apr 1;88(5):873–84. Copyright © 2003 Wiley-Liss, Inc., A Wiley Company. 34 Engineering of Functional Skeletal Tissues

Stages during Which Morphogens and Cytokines Regulate Mesencyhymal Stem Cell Differentiation

MSC Recruitment (PTHrP, BMP, TNF-α Family) MSC Commitment (BMP, PTHrP, VEGF) Proliferative Expansioon Cell Survival Apoptosis (BMP, PTHrP, VEGF, Wnts, IGF) (VEGF, FGF, BMPs, IGF) (TNF-α Family) Enhancement of Differentiated Function Matrix Matrix Production and Mineralization Maturation

CollagenII Aggrecan (PG) APase Collagen X Sox 9 PTHrP (R) Collagen IX BSP Fas Mineral Deposition

Mesenchymal Stem Cell Differentiation (MSC) Committed Progenitor Cell Collagen I AlkPhos Osteocalcin Runx2 TCF-b1 BSP Mineral deposition Ostrix Osteopontin Collagen Myoblast Adipoblast Osteoblast Tendon Fibroblast Chondroblast Osteoblast Preosteoblast Mature Osteocyte Osteoblast

Figure 2.2. Schematic summary of the lineage progression of mesenchymal stem-cell (MSC) differentiation. Upper panel: Mul- tiple stages of the life cycle of an MSC. The morphogenetic regulators of each stage are in parentheses. Lower panel: The separate stages of each of the major anabolic skeletal cell lineages are indicated with known markers that define each stage of their lineage progression. PTHrP, parathyroid hormone-related peptide; BMP, bone morphogenetic protein; TNF, tumor necrosis factor; VEGF, vascular endothelial growth factor; IGF, insulin-like growth factor; FGF, fibroblast growth factor; PG, large proteoglycan; BSP, bone sialoprotein; TGF, transforming growth factor.

Restoration of the original anatomic geome- able relevance, because the infl ammatory try of the tissue is an important aspect of bone signals spread out from the point of origin of repair. For this to occur there must be some the injury [16, 51, 54]. Data that support the role relationship between the original structure of infl ammatory cytokines play in the initiation the tissue and the gradients of the morphogens of skeletal tissue repair come from studies that promote the developmental process and showing that in the absence of TNF-α signaling the characteristics of the injury. One obvious in receptor-null animals, the callus does not functional role must be attributed to the signals develop symmetrically around the fracture that initiate and establish the symmetry of line. The absence of TNF-α signaling also leads bone repair around the fracture line. These to a delay in intramembranous and endochon- signals may be thought of as arising from the dral bone formation. Thus, TNF-α signaling marrow or from the injured cortical bone facilitates the repair process, perhaps by stimu- matrix. In this connection, how the injury lating MSC recruitment or differentiation infl uences tissue responses may have consider- [68]. Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 35

The structural geometry of the callus may skeletal-cell lineages. Of particular interest is also depend on the muscular anatomy or vas- the fact that some factors act at several stages cularization of the tissue and on the biome- during skeletal-cell lineage progression. For chanical environment at the site of injury. The example, BMPs not only are associated with latter seems to be particularly important. When MSC lineage commitment but also are involved bending and shear loading were introduced at in cellular expansion. In contrast, the scope an osteotomy site, osteogenesis was favored of morphogens such as VEGF and the Wnts over chondrogenesis [42]. Other studies have appears to be more restricted, with their pre- similarly shown that mechanical instability dominant effect on proliferative lineage expan- leads to persistence of cartilage tissue at the sion or survival. Members of the TNF-α family, fracture site. This involves up-regulation of which are part of the immune response to molecular signals such as Ihh that regulate injury, regulate the initial stages of MSC recruit- chondrogenesis [124]. How morphogenetic ment and cell survival during the infl ammatory fi elds are established and how biomechanical stage and re-emerge at the end of the MSC cycle factors direct tissue differentiation and the to control apoptosis during tissue remodeling. geometry of the regenerative process are ques- Finally, factors such as the FGFs control the rate tions of considerable importance, because the or timing of entry and exit of committed cells answers may identify the signal molecules and during their period of proliferative expansion. relate them to the origins of MSCs. Defi ning The functional contribution of specifi c cyto- how the morphogenetic fi elds are established kines and morphogens during fracture healing also has clinical importance, since the thera- is presented in Fig. 2.3. These factors are peutic responses to bioactive factors may expressed during different phases of fracture depend on whether they are correctly directed healing and therefore may vary in the roles to the morphogenetic fi eld. they play during healing. For example, TGF-β2, TGF-β3, and GDF-5 show peak mRNA expres- sion during chondrogenic differentiation and as the endochondral phases develop. This sug- 2.4 Bone Repair Is gests that the two factors are functionally Dependent upon restricted to the periods in which chondrogen- esis takes place. Multiple Cellular and Understanding the temporal pattern and Molecular Signals molecular nature of the factors as they are expressed during bone healing can allow targeting and modifi cation of their actions to The cellular and molecular processes that lead to better fracture healing. Knowing the govern bone repair after injury have many fea- spatial nature of the morphogenetic fi elds tures that are similar to what occurs in a growth during the temporal processes of fracture plate during embryonic and postnatal skeletal healing has clinical importance because the development. As reviewed earlier, fracture therapeutic responses to bioactive factors may healing involves several stages and is mediated be infl uenced by the moment in time when they by very different biological processes. Figure contact the correct morphogenetic fi eld. Such 2.2 presents the stages and progression of MSC knowledge will help to develop therapeutic differentiation into cartilage and bone as the agents to treat osteoporosis and can equally skeleton is formed. well be applied to the development of therap- Figure 2.2 also shows the stages at which eutic agents that promote bone formation. various morphogens and cytokines become Table 2.1 lists the biological processes and active and regulate MSC and skeletal-cell dif- approaches that can be modifi ed in coupled ferentiation. In addition the fi gure lists the spe- bone remodeling, either to impede bone loss or cifi c transcription factors (Runx 2, Osterix, and to promote bone regeneration. The table also Sox 9 [117]) involved in lineage commitment lists approaches that could enhance the rate or and identifi es stage-specifi c markers for the two quality of bone healing. 36 Engineering of Functional Skeletal Tissues

STAGES OF FRACTURE REPAIR Initial Injury Endochondral Formation Primary Bone Formation Secondary Bone Formation Inflammation Periosteal Response Cartilage Resorption Coupled Remodeling RELATIVE TIMES

Cytokines M-CSF IL-a IL-1b IL-6 IL-11 RANKL OPG INF-γ TNF-α TRAIL VEG1

Morphogens TGF-b1 TGF-b2 TGF-b3 BMP-2 BMP-3 BMP-4 BMP-5 BMP-6 BMP-7 BMP-8a GDF-1 GDF-5 GDF-8 GDF-10 Ihh PTHrP Wtn 4 Wtn 5b Wtn 10b Wtn 11

Proteases MMP-2 MMP-8 MMP-9 MMP-13 MMMP-14 Angiogenic VEGF A VEGF B VEGF C VEGF D Ang 1 Ang 2 Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 37

Figure 2.3. Schematic summary of the stages of fracture repair and their associated molecular processes. The relative temporal aspects of each of the stages of the fracture healing process are denoted by basic geometric shapes that also connote the relative intensity of the molecular processes that define each of the stages. The relative levels of expression of various mRNAs that have been examined in our laboratories are denoted by three line widths. The levels of expression are in percent over baseline for each and are not comparable for the various mRNAs. Data for expression levels for the proinflammatory cytokines and the extracellullar matrix (ECM) mRNAs are from Kon et al., 2001 [118]; data for TGF-α family members are from Cho et al., 2002 [38]; data for prote- ases and angiogenic factors from are from Lehmann et al., 2002 [127]; and data for Cox2 are from Gerstenfeld et al., 2002 [70]. Data pertaining to Ihh and iNOs expression are unpublished. M-CSF, macrophage colony-stimulating factor; IL, interleukin; RANKL, RANK ligand; OPG, osteoprotegrin; INF, interferon; TNF, tumor necrosis factor; VEG1, xxx; TGF, transforming growth factor; BMP, bone morphogenetic protein; GDF, growth and differentiation factor; Ihh, Indian hedgehog; PTHrP, parathyroid hormone-related peptide; MMP, matrix metalloproteinase; VEGF, vascular endothelial growth factor; Ang, angiopoietin. Reproduced with permission from Gerstenfeld LC, Cullinane DM, Barnes GL, et al. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem. 2003 Apr 1;88(5):873–84. Copyright © 2003 Wiley-Liss, Inc., A Wiley Company.

Table 2.1. Comparison of strategies in the development of therapeutic agents to treat osteoporosis versus fracture and bone repair A. Stages of fracture repair and strategies to enhance fracture repair Initial injury Endochondral formation Primary bone formation Secondary bone formation Inflammation Periosteal response Cartilage resorption Coupled remodeling Factors that promote Increase ratio of bone/ Factors that change rates of Factors that enhance coupled stem-cell recruitment cartilage differentiation endochondral remodeling bone formation (TNF family) (PTH, BMPs) (FGFs, Wnts, PTH) (TNF family) B. Stages of coupled remodeling and strategies to enhance bone mass Activation Diminish numbers of osteoclasts (TNF family) Resorption Diminish osteoclast activity/increase rate of osteoclast turnover (TNF family) Formation Increase osteoblast numbers/osteoblast activity (BMPs, PTH, Wnts)

2.5 Future Perspectives vidual factors has had mixed success in pro- moting bone healing. Regaining biomechanical on Therapeutic Uses of competency more quickly is even more compli- cated than promoting stem-cell differentiation. Morphogenetic Factors Biomechanical competency involves many factors, including the restoration of the mate- Reduction of the morbidity associated with rial properties of the tissue and of appropriate some 5% to 10% of fractures and improvement skeletal-tissue geometry. At the same time, it of healing after osteotomies, arthrodeses, will be necessary to defi ne appropriate modali- spinal fusions, and other reconstructive ortho- ties for using repair-promoting factors and to pedic procedures depend on better understand- identify when, where, and how long the factors ing of the biology of fracture and bone healing should be applied. Because many factors, once (170). As discussed above, multiple morphoge- they activate receptors, utilize overlapping netic factors regulate normal skeletal develop- signal-transduction pathways to mediate intra- ment, but it is not clear how they function in cellular effects, signal pathways need to be postnatal healing. Many factors act coopera- identifi ed in the hope of making optimal use of tively or even antagonistically at different the small-molecule pharmaceuticals that are stages of bone development. Single use of indi- being developed. 38 Engineering of Functional Skeletal Tissues

(VEGF) is mediated via the VEGF receptor fl t-1. References Blood 87:3336–3343. 16. Barnes GL, Kostenuik PJ, Gerstenfeld LC, Einhorn TA (1999) Growth factor regulation of fracture repair. 1. Abbas S, Zhang YH, Clohisy JC, Abu-Amer Y (2003) J Bone Miner Res 14:1805–1815. Tumor necrosis factor-alpha inhibits pre-osteoblast 17. Bazzoni F, Beutler B (1996) The tumor necrosis factor differentiation through its type-1 receptor. Cytokine ligand and receptor family. N Engl J Med 334: 22:33–41. 1717–1725. 2. Abou-Samra AB, Juppner H, Force T, Freeman MW, 18. Bellus GA, McIntosh I, Smith EA, Aylsworth AS, Kong XF, Schipani E, Urena P, Richards J, Bonventre Kaitila I, Horton WA, Greenhaw GA, Hecht JT, Fran- JV, Potts JT Jr, Kronenberg HM, Segre GV (1992) comano CA (1995) A recurrent mutation in the tyro- Expression cloning of a common receptor for parathy- sine kinase domain of fi broblast growth factor roid hormone and parathyroid hormone-related receptor 3 causes hypochondroplasia. Nat Genet peptide from rat osteoblast-like cells: a single receptor 10:357–359. stimulates intracellular accumulation of both cAMP 19. Bhardwaj A, Aggarwal BB (2003) Receptor-mediated and inositol trisphosphates and increases in trocellu- choreography of life and death. J Clin Immunol lar free calcium. Proc Natl Acad Sci USA 89:2732– 23:317–332. 2736. 20. Bianco P, Riminucci M, Gronthos S, Robey PG (2001) 3. Adams CS, Shapiro IM (2002) The fate of the termi- Bone marrow stromal stem cells: nature, biology, and nally differentiated chondrocyte: evidence for micro- potential applications. Stem Cells 19:180–192. environmental regulation of chondrocyte apoptosis. 21. Bitzer M, von Gersdorff G, Liang D, Dominguez- Crit Rev Oral Biol Med 13:465–473. Rosales A, Beg AA, Rojkind M, Bottinger EP (2000) 4. Aigner T, Kim H (2002) Apoptosis and cellular vital- A mechanism of suppression of TGF-beta/SMAD sig- ity: issues in osteoarthritic cartilage degeneration. naling by NFkappa B/RelA. Genes Dev 14:187–197. Arthritis Rheum 46:1986–1996. 22. Blomstrand S, Claesson I, Save-Soderbergh J (1985) 5. Aizawa T, Kokubun S, Tanaka Y (1997) Apoptosis A case of lethal congenital dwarfi sm with accelerated and proliferation of growth plate chondrocytes in skeletal maturation. Pediatr Radiol 15:141–143. rabbits. J Bone Joint Surg Br 79B:483–486. 23. Bluethmann H (1998) Physiological, immunological, 6. Aizawa T, Kon T, Einhorn TA, Gerstenfeld LC and pathological functions of tumor necrosis factor (2001) Induction of apoptosis in chondrocytes by (TNF) revealed by TNF receptor-defi cient mice. In: tumor necrosis factor-alpha. J Orthop Res 19:785– Durum S, Muegge K, eds. Cytokine Knockouts. 796. Totowa: Humana Press, pp 69–87. 7. Andreassen TT, Ejersted C, Oxlund H (1999) Intermit- 24. Boland GM, Perkins G, Hall DJ, Tuan RS (2004) Wnt tent parathyroid hormone (1–34) treatment increases 3a promotes proliferation and suppresses osteogenic callus formation and mechanical strength of healing differentiation of adult human mesenchymal stem rat fractures. J Bone Miner Res 14:960–968. cells. J Cell Biochem 93:1210–1230. 8. Angel P, Imagawa M, Chiu R, Stein B, Imbra RJ, 25. Bostrom MP, Gamradt SC, Asnis P, Vickery BH, Hill Rahmsdorf HJ, Jonat C, Herrlich P, Karin M (1987) E, Avnur Z, Waters RV (2000) Parathyroid hormone- Phorbol ester-inducible genes contain a common cis related protein protein analog RS-66271 is an effec- element recognized by a TPA-modulated trans- tive therapy for impaired bone healing in rabbits on acting factor. Cell 49:729–739. corticosteroid therapy. Bone 2:437–442. 9. Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, 26. Bostrom MP, Lane JM, Berberian WS, Missri AA, Takubo K, Ito K, Koh GY, Suda T (2004) Tie2/ Tomin E, Weiland A, Doty SB, Glaser D, Rosen VM angiopoietin-1 signaling regulates hematopoietic (1995) Immunolocalization and expression of bone stem cell quiescence in the bone marrow niche. Cell morphogenetic protein 2 and 4 in fracture healing. J 118:149–161. Orthop Res 13:357–367. 10. Aronson J, Harp JH, Walker CW, Dalrymple GV 27. Bouletreau PJ, Warren SM, Spector JA, Peled ZM, (1990) Blood fl ow, bone formation and mineraliza- Gerrets RP, Greenwald JA, Longaker MT (2002) tion during . Trans Orthop Hypoxia and VEGF up-regulate BMP-2 mRNA and Res Soc 15:589–594. protein expression in microvascular endothelial 11. Attisano L, Labbe E (2004) TGF-beta and Wnt pathway cells: implications for fracture healing. Plast Recon- cross-talk. Cancer Metastasis Rev 23:53–61. str Surg 109:2384–2397. 12. Babij P, Zhao W, Small C, Kharode Y, Yaworsky PJ, 28. Broadus AE, Stewart AF (1994) Parathyroid hormone- Bouxsein ML, Reddy PS, Bodine PV, Robinson JA, related protein: structure, processing, and physio- Bhat B, Marzolf J, Moran RA, Bex F (2003) High bone logical actions. In: Bilezikian JP, Levine MA, Marcus mass in mice expressing a mutant LRP5 gene. J Bone R, eds. The Parathyroids: Basic and Clinical Con- Miner Res 18:960–974. cepts. New York: Raven Press, pp 259–294. 13. Babu P (1977) Early developmental subdivisions of 29. Buckwalter JA, Einhorn TA, Marsh LJ (2001) Bone and the wing disk in Drosophila. Mol Gen Genet 151: joint healing. In: Bucholz RW, Heckman JD, eds. 289–294. Rockwood and Green’s Fractures in Adults. Philadel- 14. Baker NE (1988) Localization of transcripts from the phia: Lippincott, Williams, and Wilkins, pp 245–271. wingless gene in whole Drosophila embryos. Devel- 30. Calvi LM, Adams GB, Weibrecht KW, Weber JM, opment 103:289–298. Olson DP, Knight MC, Martin RP, Schipani E, Divieti 15. Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani P, Bringhurst FR, Milner LA, Kronenberg HM, A, Marme D (1996) Migration of human monocytes Scadden DT (2003) Osteoblastic cells regulate the in response to vascular endothelial growth factor hematopoietic stem cell niche. Nature 425:841–846. Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 39

31. Candeliere GA, Glorieux FH, Prud’homme J, 45. Dealy CN, Roth A, Ferrari D, Brown AM, Kosher RA St-Arnaud R (1995) Increased expression of the c-fos (1993) Wnt-5a and Wnt-7a are expressed in the proto-ocogene in bone from patients with fi brous developing chick limb bud in a manner suggesting dysplasia. N Engl J Med 332:1546–1551. roles in pattern formation along the proximo 32. Carano RA, Filvaroff EH (2003) Angiogenesis and distal and dorsoventral axes. Mech Dev 43:175– bone repair. Drug Discov Today 8:980–989. 186. 33. Carlevaro MF, Cermelli S, Cancedda R, Descalzi 46. Deckers MM, Karperien M, van der Bent C, Yamashita Cancedda F (2000) Vascular endothelial growth T, Papapoulos SE, Lowik CW (2000) Expression of factor (VEGF) in cartilage neovascularization and vascular endothelial growth factors and their recep- chondrocyte differentiation: auto-paracrine role tors during osteoblast differentiation. Endocrinol- during endochondral bone formation. J Cell Sci ogy 141:1667–1674. 113:59–69. 47. Deckers MM, van Bezooijen RL, van der Horst G, 34. Carvalho RS, Einhorn TA, Lehmann W, Edgar C, Hoogendam J, van Der Bent C, Papapoulos SE, Lowik Al-Yamani A, Apazidis A, Pacicca D, Clemens TL, CW (2002) Bone morphogenetic proteins stimulate Gerstenfeld LC (2004) The role of angiogenesis in a angiogenesis through osteoblast-derived vascular murine tibial model of distraction osteogenesis. endothelial growth factor A. Endocrinology 143: Bone 34:849–861. 1545–1553. 35. Celeste AJ, Iannazzi JA, Taylor RC, Hewick RM, 48. Dempster DW, Cosman F, Kurland ES, Zhou H, Rosen V, Wang EA, Wozney JM (1990) Identifi cation Nieves J, Woelfert L, Shane E, Plavetic K, Muller R, of transforming growth factor beta family members Bilezikian J, Lindsay R (2001) Effects of daily treat- present in bone-inductive protein purifi ed from ment with parathyroid hormone on bone microar- bovine bone. Proc Natl Acad Sci 87:9843–9847. chitecture and turnover in patients with osteoporosis: 36. Chen S, Guttridge DC, Tang E, Shi S, Guan K, Wang a paired biopsy study. J Bone Miner Res 16:1846– CY (2001) Suppression of tumor necrosis factor- 1853. mediated apoptosis by nuclear factor kappaB- 49. Dosch R, Gawantka V, Delius H, Blumenstock C, independent bone morphogenetic protein/Smad Niehrs C (1997) BMP-4 acts as a morphogen in dor- signaling. J Biol Chem 276:39259–39263. soventral mesoderm patterning in Xenopus. Devel- 37. Childs LM, Paschalis EP, Xing L, Dougall WC, Ander- opment 124:2325–2334. son D, Boskey AL, Puzas JE, Rosier RN, O’Keefe RJ, 50. Dunstan CR, Boyce R, Boyce BF, Garrett IR, Izbicka Boyce BF, Schwarz EM (2002) In vivo RANK signal- E, Burgess WH, Mundy GR (1999) Systemic adminis- ing blockade using the receptor activator of NF- tration of acidic fi broblast growth factor (FGF-1) kappaB:Fc effectively prevents and ameliorates wear prevents bone loss and increases new bone formation debris-induced via osteoclast depletion in ovariectomized rats. J Bone Miner Res 14:953– without inhibiting osteogenesis. J Bone Miner Res 959. 17:192–199. 51. Einhorn TA (1995) Enhancement of fracture healing. 38. Cho T-J, Gerstenfeld LC, Einhorn TA (2002) J Bone Joint Surg 77:940–956. Differential temporal expression of members of 52. Einhorn T, Lee C (2001) Bone regeneration: new fi nd- the transforming growth factor beta superfamily ings and potential clinical applications. J Am Acad during murine fracture healing. J Bone Miner Res Orthop Surg 9:157–165. 17:513–520. 53. Einhorn TA, Majeska RJ, Mohaideen A, Kagel EM, 39. Cho T-J, Lehmann W, Edgar C, Sadeghi C, Hou A, Bouxsein ML, Turek TJ, Wozney JM (2003) A single Einhorn TA, Gerstenfeld LC (2003) Tumor necrosis percutaneous injection of recombinant human bone factor alpha activation of the apoptotic cascade morphogenetic protein-2 accelerates fracture repair. in murine articular chondrocytes is associated J Bone Joint Surg Am 85-A:1425–1435. with the induction of metalloproteinases and specifi c 54. Einhorn TA, Majeska RJ, Rush EB, Levine PM, pro-resorptive factors. Arthritis Rheum 48:2845– Horowitz MC (1995) The expression of cytokine 2854. activity by fracture callus. J Bone Miner Res 10: 40. Clauss IM, Gravallese EM, Darling JM (1993) In situ 1272–1281. hybridization studies suggest a role for the basic 55. Eugster H-P, Muller M, LeHir M, Ryffel B (1998) region-leucine zipper protein hXBP-1 in exocrine Immunodefi ciency of tumor necrosis factor and gland and skeletal development during mouse lymophotoxin-alpha double defi cient mice. In: embryogenesis. Dev Dyn 197:146–156. Durum S, Muegge K, editors. Cytokine Knockouts. 41. Constantinides PG, Taylor SM, Jones PA (1978) Phe- Totowa: Humana Press, pp 103–118. notypic conversion of cultured mouse embryo cells 56. Farnum CE, Willsman NJ (1989) Cellular turnover at by aza pyrimidine nucleosides. Dev Biol 66:57–71. the chondro-osseous junction of growth plate carti- 42. Cullinane DM, Fredrick A, Eisenberg SR, Pacicca D, lage: analysis by serial sections at the light micro- Elman MV, Lee C, Salisbury K, Gerstenfeld LC, scopical level. J Orthop Res 7:654–666. Einhorn TA (2002) Induction of a neoarthrosis by 57. Feng JQ, Xing L, Zhang JH, Zhao M, Horn D, Chan J, precisely controlled motion in an experimental mid- Boyce BF, Harris SE, Mundy GR, Chen D (2003) femoral defect. J Orthop Res 20:579–586. NFkappaB specifi cally activates BMP-2 gene expres- 43. Danial NN, Korsmeyer SJ (2004) Cell death: critical sion in growth plate chondrocytes in vivo and control points. Cell 116:205–219. in a chondrocyte cell line in vitro. J Biol Chem 44. De Robertis EM, Larrain J, Oelgeschlager M, Wessely 278:29130–29135. O (2000) The establishment of Spemann’s organizer 58. Ferguson C, Alpern E, Miclau T, Helms JA (1999) and patterning of the vertebrate embryo. Nat Rev Does adult fracture repair recapitulate embryonic Genet 1:171–181. skeletal formation? Mech Dev 87:57–66. 40 Engineering of Functional Skeletal Tissues

59. Ferrara N (1999) Role of vascular endothelial growth Carle GF, Dallapiccola B, De Paepe A, Floege B, Half- factor in the regulation of angiogenesis. Kidney Int hide ML, Hall B, Hennekam RC, Hirose T, Jans A, 56:794–814. Juppner H, Kim CA, Keppler-Noreuil K, Kohlschuetter 60. Ferrara N (2000) VEGF: an update on biological A, LaCombe D, Lambert M, Lemyre E, Letteboer T, and therapeutic aspects. Curr Opin Biotechnol 11: Peltonen L, Ramesar RS, Romanengo M, Somer H, 617–624. Steichen-Gersdorf E, Steinmann B, Sullivan B, 61. Ferrara N (2001) Role of vascular endothelial Superti-Furga A, Swoboda W, van den Boogaard MJ, growth factor in regulation of physiological angio- Van Hul W, Vikkula M, Votruba M, Zabel B, Garcia genesis. Am J Physiol Cell Physiol 280:C1358– T, Baron R, Olsen BR, Warman ML, for the Osteopo- C1366. rosis-Pseudoglioma Syndrome Collaborative Group 62. Fitzpatrick LA, Coleman DT, Bilezikian JP (1992) (2001) LDL receptor-related protein 5 (LRP5) affects The target tissue actions of parathyroid hormone. bone accrual and eye development. Cell 107:513– In: Coe FL, Favus MJ, eds. Disorders of Bone and 523. Mineral Metabolism. New York: Raven Press, 74. Goralczyk R, Closs EI, Ruther U, Wagner EF, Strauss pp 123–147. PG, Erfl e V, Schmidt J (1990) Characterization of fos- 63. Fukuhara H, Mizuno K (1989) The infl uence of para- induced osteogenic tumours and tumour-derived thyroid hormone on the process of fracture healing. murine cell lines. Differentiation 44:122–131. Nippon Seikeigeka Gakkai Zasshi 63:100–115. 75. Gravallese EM, Pettit AR, Lee R, Madore R, Manning 64. Fukui N, Zhu Y, Maloney WJ, Clohisy J, Sandell LJ C, Tsay A, Gaspar J, Goldring MB, Goldring SR, (2003) Stimulation of BMP-2 expression by pro- Oettgen P (2003) Angiopoietin-1 is expressed in the infl ammatory cytokines IL-1 and TNF-alpha in synovium of patients with rheumatoid arthritis and normal and osteoarthritic chondrocytes. J Bone is induced by tumor necrosis factor alpha. Ann Joint Surg Am 85A:59–66. Rheum Dis 62:100–107. 65. Gardella TJ, Jüppner H (2001) Molecular properties 76. Guo J, Chung UI, Kondo H, Bringhurst FR, Kronen- of the PTH/PTHrP receptor. Trends Endocrinol berg HM (2002) The PTH/PTHrP receptor can delay Metab 12:210–217. chondrocyte hypertrophy in vivo without activating 66. Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, phospholipase C. Dev Cell 3:183–194. Ferrara N (1999) VEGF couples hypertrophic carti- 77. Habener JF (1990) Cyclic AMP response element lage remodeling, ossifi cation and angiogenesis binding proteins: a cornucopia of transcription during endochondral bone formation. Nat Med factors. Mol Endocrinol 4:1087–1094. 5:623–628. 78. Harbour ME, Gregory JW, Jenkins HR, Evans BA 67. Gerhart TN, Kirker-Head CA, Kriz MJ, Holtrop ME, (2000) Proliferative response of different human Hennig GE, Hipp J, Schelling SH, Wang E (1993) osteoblast-like cell models to proinfl ammatory cyto- Healing segmental femoral defects in sheep using kines. Pediatr Res 48:163–168. recombinant human bone morphogenetic protein. 79. Haridas V, Shrivastava A, Su J, Yu GL, Ni J, Liu D, Clin Orthop 293:317–326. Chen SF, Ni Y, Ruben SM, Gentz R, Aggarwal BB 68. Gerstenfeld LC, Cho T-J, Kon T, Aizawa T, Cruceta J, (1999) VEGI, a new member of the TNF family acti- Graves BD, Einhorn TA (2001) Impaired intramem- vates nuclear factor-kappa B and c-Jun N-terminal branous bone formation during bone repair in the kinase and modulates cell growth. Oncogene absence of tumor necrosis factor-alpha signaling. 18:6496–6504. Cells Tissues Organs 169:285–294. 80. Harper J, Gerstenfeld LC, Klagsbrun M (2001) Neu- 69. Gerstenfeld LC, Cho T-J, Kon T, Aizawa T, Tsay A, ropilin-1 expression in osteogenic cells: down-regu- Fitch J, Barnes GL, Graves DT, Einhorn TA (2003) lation during differentiation of osteoblasts into Impaired fracture healing in the absence of TNF- osteocytes. J Cell Biochem 81:82–92. alpha signaling: the role of TNF-alpha in endochon- 81. Hartmann C, Tabin CJ (2000) Dual roles of Wnt sig- dral cartilage resorption. J Bone Miner Res naling during chondrogenesis in the chicken limb. 18:1584–1592. Development 127:3141–3159. 70. Gerstenfeld LC, Cruceta J, Shea CM, Sampath K, 82. Harty M, Neff AW, King MW, Mescher AL (2003) Barnes GL, Einhorn TA (2002) Chondrocytes provide Regeneration or scarring: an immunologic perspec- morphogenic signals that selectively induce osteo- tive. Dev Dyn 226:268–279. genic differentiation of mesenchymal stem cells. 83. Hashimoto S, Setareh M, Ochs R, Lotz M (1997) Fas/ J Bone Miner Res 17:221–230. Fas ligand expression and induction of apoptosis in 71. Gerstenfeld LC, Shapiro FD (1996) Expression of chondrocytes. Arthritis Rheum 40:1749–1755. bone-specifi c genes by hypertrophic chondrocytes: 84. Hausman MR, Schaffl er MB, Majeska RJ (2001) Pre- implication of the complex functions of the hyper- vention of fracture healing in rats by an inhibitor of trophic chondrocyte during endochondral bone angiogenesis. Bone 29:560–564. development. J Cell Biochem 62:1–9. 85. Hock JM, Gera I (1992) Effects of continuous and 72. Gibson GJ, Kohler WJ, Schaffl er MB (1995) Chondro- intermittent administration and inhibition of resorp- cyte apoptosis in endochondral ossifi cation of chick tion on the anabolic response of bone to parathyroid sterna. Dev Dyn 203:468–476. hormone. J Bone Miner Res 7:65–72. 73. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman 86. Hofmann C, Luo G, Balling R, Karsenty G (1996) S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev Analysis of limb patterning in BMP-7-defi cient mice. D, Zacharin M, Oexle K, Marcelino J, Suwairi W, Dev Genet 19:43–50. Heeger S, Sabatakos G, Apte S, Adkins WN, Allgrove 87. Holash J, Wiegand SJ, Yancopoulos GD (1999) New J, Arslan-Kirchner M, Batch JA, Beighton P, Black model of tumor angiogenesis: dynamic balance GC, Boles RG, Boon LM, Borrone C, Brunner HG, between vessel regression and growth mediated Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 41

by angiopoietins and VEGF. Oncogene 18:5356– thyroid hormone-related peptide gene. Genes Dev 5362. 8:277–289. 88. Holzer G, Majeska RJ, Lundy MW, Hartke JR, Einhorn 103. Karp SJ, Schipani E, St-Jacques B, Hunzelman J, Kro- TA (1999) Parathyroid hormone enhances fracture nenberg H, McMahon AP (2000) Indian hedgehog healing. Clin Orthop 366:258–263. coordinates endochondral bone growth and mor- 89. Horner A, Bord S, Kelsall AW, Coleman N, Compston phogenesis via parathyroid hormone related-protein- JE (2001) Tie2 ligands angiopoietin-1 and angiopoi- dependent and -independent pathways. Development etin-2 are coexpressed with vascular endothelial cell 127:543–548. growth factor in growing human bone. Bone 28:65– 104. Katagiri T, Yamaguchi A, Komaki M, Abe E, Taka- 71. hashi N, Ikeda T, Rosen V, Wozney JM, Fujisawa- 90. Hunziker EB, Schenk RK, Cruz-Orive LM (1987) Sehara A, Suda T (1994) Bone morphogenetic Quantitation of chondrocyte performance in growth- protein-2 converts the differentiation pathway of plate cartilage during longitudinal bone growth. J C2C12 myoblasts into the osteoblast lineage. J Cell Bone Joint Surg Am 69:162–173. Biol 127:1755–1766. 91. Ikeya M, Takada S (2001) Wnt-3a is required for 105. Katavic V, Lukic IK, Kovacic N, Grcevic D, Lorenzo somite specifi cation along the anteroposterior axis of JA, Marusic A (2003) Increased bone mass is a the mouse embryo and for regulation of cdx-1 expres- part of the generalized lymphoproliferative disorder sion. Mech Dev 103:27–33. phenotype in the mouse. J Immunol 170:1540– 92. Iwata H, Sakano S, Itoh T, Bauer TW (2002) Demin- 1547. eralized bone matrix and native bone morphogenetic 106. Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, protein in orthopaedic surgery. Clin Orthop Glass DA 2nd, Hartmann C, Li L, Hwang TH, Brayton 395:99–109. CF, Lang RA, Karsenty G, Chan L (2002) Cbfa1-inde- 93. Jabs EW, Li X, Scott AF, Meyers G, Chen W, Eccles M, pendent decrease in osteoblast proliferation, osteo- Mao JI, Charnas LR, Jackson CE, Jaye M (1994) penia, and persistent embryonic eye vascularization Jackson-Weiss and Crouzon syndromes are allelic in mice defi cient in Lrp5, a Wnt coreceptor. J Cell Biol with mutations in fi broblast growth factor receptor 157:303–314. 2. Nat Genet 8:275–279. 107. Kawabata M, Imamura T, Miyazono K (1998) Signal 94. Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP transduction by bone morphogenetic proteins. Cyto- (1997) Osteogenic differentiation of purifi ed, culture- kine Growth Factor Rev 9:49–61. expanded human mesenchymal stem cells in vitro. 108. Kawakami Y, Capdevila J, Buscher D, Itoh T, J Cell Biochem 64:295–312. Rodriguez Esteban C, Izpisua Belmonte JC (2001) 95. Jansen M (1934) Über atypische Chondrodystophie WNT signals control FGF-dependent limb initiation (Achondroplasie) und über eine noch nicht bes- and AER induction in the chick embryo. Cell chriebene angeborene Wachstumsstörung des Kno- 104:891–900. chensystems: Metaphysäre Dysostosis. Zeitschr 109. Kawakami Y, Wada N, Nishimatsu SI, Ishikawa T, Orthop Chir 61:253–286. Noji S, Nohno T (1999) Involvement of Wnt-5a in 96. Jingushi S, Urabe K, Okazaki K, Hirata G, Sakai A, chondrogenic pattern formation in the chick limb Ikenoue T, Iwamoto Y (2002) Intramuscular bone bud. Dev Growth Differ 41:29–40. induction by human recombinant bone morphoge- 110. Kengaku M, Capdevila J, Rodriguez-Esteban C, De La netic protein-2 with beta-tricalcium phosphate as a Pena J, Johnson RL, Belmonte JC, Tabin CJ (1998) carrier: in vivo bone banking for muscle-pedicle Distinct WNT pathways regulating AER formation autograft. J Orthop Sci 7:490–494. and dorsoventral polarity in the chick limb bud. 97. Jobert AS, Zhang P, Couvineau A, Bonaventure J, Science 280:1274–1277. Roume J, Le Merrer M, Silve C (1998) Absence of 111. Kiener PA, Davis PM, Starling GC, Mehlin C, Kle- functional receptors for parathyroid hormone and banoff SJ, Ledbetter JA, Liles WC (1997) Differential parathyroid hormone-related peptide in Blomstrand induction of apoptosis by Fas-Fas ligand interactions chondrodysplasia. J Clin Invest 102:34–40. in human monocytes and macrophages. J Exp Med 98. Jones PF (2003) Not just angiogenesis—wider roles 185:1511–1516. for the angiopoietins. J Pathol 201:515–527. 112. Kim HW, Jahng JS (1999) Effect of intermittent 99. Ju ST, Panka DJ, Cui H, Ettinger R, el-Khatib M, administration of parathyroid hormone on fracture Sherr DH, Stanger BZ, Marshak-Rothstein A (1995) healing in ovariectomized rats. Iowa Orthop J Fas (CD95)/FasL interactions required for pro- 19:71–77. grammed cell death after T-cell activation. Nature 113. Kim KS, Tinti C, Song B, Cubells JF, Joh TH (1994) 373:444–448. Cyclic AMP-dependent protein kinase regulates 100. Kaku M, Kohno S, Kawata T, Fujita I, Tokimasa C, basal and cyclic AMP-stimulated but not phorbol Tsutsui K, Tanne K (2001) Effects of vascular ester-stimulated transcription of the tyrosine endothelial growth factor on osteoclast induction hydroxylase gene. J Neurochem 63:834–842. during tooth movement in mice. J Dent Res 80:1880– 114. Kim N, Odgren PR, Kim DK, Marks SC Jr, Choi Y 1883. (2000) Diverse roles of the tumor necrosis factor 101. Kanzler B, Foreman RK, Labosky PA, Mallo M (2000) family member TRANCE in skeletal physiology BMP signaling is essential for development of skele- revealed by TRANCE defi ciency and partial rescue togenic and neurogenic cranial neural crest. Devel- by a lymphocyte-expressed TRANCE transgene. opment 127:1095–1104. Proc Natl Acad Sci USA 97:10905–10910. 102. Karaplis AC, Luz A, Glowacki J, Bronson RT, Tybule- 115. Kimble RB, Bain S, Pacifi ci R (1997) The functional wicz VL, Kronenberg HM, Mulligan RC (1994) Lethal block of TNF but not IL-6 prevents bone loss in ovari- skeletal dysplasia from targeted disruption of para- ectomized mice. J Bone Miner Res 12:935–941. 42 Engineering of Functional Skeletal Tissues

116. Kimmel DB, Bozzato RP, Kronis KA, Coble T, 130. Liekens S, De Clercq E, Neyts J (2001) Angiogenesis: Sindrey D, Kwong P, Recker RR (1993) The effect of regulators and clinical applications. Biochem Phar- recombinant human (1–84) or synthetic human (1– macol 61:253–270. 34) parathyroid hormone on the skeleton of adult 131. Liou H-C, Boothby MR, Finn PW, Davidon R, Nabavi osteopenic ovariectomized rats. Endocrinology N, Zeleznik-Le NJ, Ting JP, Glimcher LH (1990) A 132:1577–1584. new member of the leucine zipper class of proteins 117. Kobayashi T, Kronenberg H (2005) Minireview: tran- that binds to the HLA DR alpha promoter. Science scriptional regulation in development of bone. Endo- 247:1581–1584. crinology 146:1012–1017. 132. Liou H-C, Boothby MR, Glimcher LH (1988) Distinct 118. Kon T, Cho T-J, Aizawa T, Yamazaki M, Nooh N, cloned class II MHC DNA binding proteins recognize Graves D, Gerstenfeld LC, Einhorn TA (2001) Expres- the X transcription element. Science 242:69–71. sion of osteoprotegerin, receptor activator of NF- 133. Liu F, Ventura F, Doody J, Massague J (1995) Human kappaB ligand (osteoprotegerin ligand) and related type II receptor for bone morphogenic proteins proinfl ammatory cytokines during fracture healing. (BMPs): extension of the two-kinase receptor model J Bone Miner Res 16:1004–1014. to the BMPs. Mol Cell Biol 15:3479–3486. 119. Kostiainen K (1996) Recombinant bone morphoge- 134. Liu X, Nagarajan RP, Vale W, Chen Y (2002) Phos- netic proteins: cloning and molecular structures phorylation regulation of the interaction between (Chapter 4). In: Lindholm TS, ed. Bone Morphoge- Smad7 and activin type I receptor. Fed Euro Biochem netic Proteins: Biology, Biochemistry, and Recon- Soc Lett 519:93–98. structive Surgery. Austin: RG Landes Company and 135. Lobov IB, Brooks PC, Lang RA (2002) Angiopoietin-2 Academic Press, pp 39–46. displays VEGF-dependent modulation of capillary 120. Kronenberg HM (2003) Developmental regulation of structure and endothelial cell survival in vivo. Proc the growth plate. Nature 423:332–336. Natl Acad Sci USA 99:11205–11210. 121. Kucharczak J, Simmons MJ, Fan Y, Gelinas C (2003) 136. Long F, Zhang XM, Karp S, Yang Y, McMahon AP To be, or not to be: NFkappaB is the answer—role of (2001) Genetic manipulation of hedgehog signaling Rel/NFkappaB in the regulation of apoptosis. Onco- in the endochondral skeleton reveals a direct role in gene 22:8961–8982. the regulation of chondrocyte proliferation. Devel- 122. Lanske B, Karaplis AC, Lee K, Luz A, Vortkamp A, opment 128:5099–5108. Pirro A, Karperien M, Defi ze LH, Ho C, Mulligan RC, 137. Lories RJ, Derese I, Ceuppens JL, Luyten FP (2003) Abou-Samra AB, Juppner H, Segre GV, Kronenberg Bone morphogenetic proteins 2 and 6, expressed in HM (1996) PTH/PTHrP receptor in early develop- arthritic synovium, are regulated by proinfl amma- ment and Indian hedgehog-regulated bone growth. tory cytokines and differentially modulate fi bro- Science 273:663–666. blast-like synoviocyte apoptosis. Arthritis Rheum 123. Lawson KA, Dunn NR, Roelen BA, Zeinstra LM, 48:2807–2818. Davis AM, Wright CV, Korving JP, Hogan BL (1999) 138. Luo G, Hofmann C, Bronckers AL, Sohocki M, BMP4 is required for the generation of primordial Bradley A, Karsenty G (1995) BMP-7 is an inducer germ cells in the mouse embryo. Genes Dev 13: of nephrogenesis, and is also required for eye 66–68. development and skeletal patterning. Genes Dev 124. Le AX, Miclau T, Hu D, Helms JA (2001) Molecular 9:2808–2820. aspects of healing in stabilized and non-stabilized 139. Lyons KM, Pelton RW, Hogan BL (1990) Organo- fractures. J Orthop Res 19:78–84. genesis and pattern formation in the mouse: RNA 125. Lee KAW, Masson N (1993) Transcriptional regula- distribution patterns suggest a role for bone mor- tions by CREB and its relatives. Biochem Biophys phogenetic protein-2A (BMP-2A). Development Acta 1174:221–233. 109:833–844. 126. Lee SW, Lee HJ, Chung WT, Choi SM, Rhyu SH, 140. Maes C, Carmeliet P, Moermans K, Stockmans I, Kim DK, Kim KT, Kim JY, Kim JM, Yoo YH (2004) Smets N, Collen D, Bouillon R, Carmeliet G (2002) TRAIL induces apoptosis of chondrocytes and Impaired angiogenesis and endochondral bone for- infl uences the pathogenesis of experimentally mation in mice lacking the vascular endothelial induced rat osteoarthritis. Arthritis Rheum 50:534– growth factor isoforms VEGF164 and VEGF188. 542. Mech Dev 111:61–73. 127. Lehmann W, Edgar CM, Wang K, Cho TJ, Barnes GL, 141. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Kakar S, Graves DT, Rueger JM, Gerstenfeld LC, Wiegand SJ, Radziejewski C, Compton D, McClain J, Einhorn TA (2005) Tumor necrosis factor alpha Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato (TNF-alpha) coordinately regulates the expression TN, Yancopoulos GD (1997) Angiopoietin-2, a of specifi c matrix metalloproteinases (MMPS) and natural antagonist for Tie 2 that disrupts in vivo angiogenic factors during fracture healing. Bone angiogenesis. Science 277:55–60. 36:300–310. 142. Marie PJ (2003) Fibroblast growth factor signaling 128. Lewinson D, Maor G, Rozen N, Rabinovich I, Stahl S, controlling osteoblast differentiation. Gene 316:23– Rachmiel A (2001) Expression of vascular antigens 32. by bone cells during bone regeneration in a membra- 143. Marron MB, Hughes DP, Edge MD, Forder CL, Brindle nous bone distraction system. Histochem Cell Biol NP (2000) Evidence for heterotypic interaction 116:381–388. between the receptor tyrosine kinases TIE-1 and 129. Li G, Simpson AH, Kenwright J, Triffi tt JT (1999) TIE-2. J Biol Chem 275:39741–39746. Effect of lengthening rate on angiogenesis during 144. Martin GR (1998) The roles of FGFs in the early devel- distraction osteogenesis. J Orthop Res 17:362– opment of vertebrate limbs. Genes Dev 12:1571– 367. 1586. Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 43

145. Mayahara H, Ito T, Nagai H, Miyajima H, Tsukuda R, 159. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Taketomi S, Mizoguchi J, Kato K (1993) In vivo Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, stimulation of endosteal bone formation by basic Ish-Shalom S, Genant HK, Wang O, Mitlak BH fi broblast growth factor in rats. Growth Factors (2001) Effect of parathyroid hormone (1–34) on frac- 9:73–80. tures and bone mineral density in postmenopausal 146. Maye P, Zheng J, Li L, Wu D (2004) Multiple mecha- women with osteoporosis. N Engl J Med 344:1434– nisms for Wnt11-mediated repression of the canoni- 1441. cal Wnt signaling pathway. J Biol Chem 279:24659– 160. Nickel J, Dreyer MK, Kirsch T, Sebald W (2001) The 24665. crystal structure of the BMP-2:BMPR-IA complex 147. Mayr-Wohlfart U, Waltenberger J, Hausser H, Kessler and the generation of BMP-2 antagonists. J Bone S, Gunther KP, Dehio C, Puhl W, Brenner RE (2002) Joint Surg Am 83A(Suppl 1):S7–S14. Vascular endothelial growth factor stimulates che- 161. Nishioka K, Dennis JE, Gao J, Goldberg VM, Caplan motactic migration of primary human osteoblasts. AI (2005) Sustained Wnt protein expression in chon- Bone 30:472–477. dral constructs from mesenchymal stem cells. J Cell 148. Minina E, Kreschel C, Naski MC, Ornitz DM, Vort- Physiol 203:6–14. kamp A (2002) Interaction of FGF, Ihh/Pthlh, and 162. Nusslein-Volhard C, Wieschaus E (1980) Mutations BMP signaling integrates chondrocyte proliferation affecting segment number and polarity in Drosoph- and hypertrophic differentiation. Dev Cell 3:439– ila. Nature 287:795–801. 449. 163. Olsen NJ, Stein CM (2004) New drugs for rheumatoid 149. Miyazono K (2002) Bone morphogenetic protein arthritis. N Engl J Med 350:2167–2179. receptors and actions (Chapter 51). In: Bilezikian JP, 164. Ornitz DM, Itoh N (2001) Fibroblast growth factors. Raisz LG, Rodan GA, eds. Principles of Bone Biology. Genome Biol 2:REVIEWS3005. 2nd edition. San Diego: Academic Press, pp 929– 165. Ornitz DM, Marie PJ (2002) FGF signaling pathways 942. in endochondral and intramembranous bone devel- 150. Miyazono Y (1999) Signal transduction by bone mor- opment and human genetic disease. Genes Dev phogenetic protein receptors: functional roles of 16:1446–1465. Smad proteins. Bone 25:91–93. 166. Ozkaynak E, Rueger DC, Drier EA, Corbett C, Ridge 151. Mosekilde L, Sogaard CH, Danielsen CC, Torring O RJ, Sampath TK, Oppermann H (1990) OP-1 cDNA (1991) The anabolic effects of human parathyroid encodes an osteogenic protein in the TFG-beta hormone (hPTH) on rat vertebral body mass are also family. EMBO J 9:2085–2093. refl ected in the quality of bone, assessed by biome- 167. Park YS, Kim NH, Jo I (2003) Hypoxia and vascular chanical testing: a comparison study between hPTH- endothelial growth factor acutely up-regulate angio- (1–34) and hPTH-(1–84). Endocrinology 129:421– poietin-1 and Tie2 mRNA in bovine retinal pericytes. 428. Microvasc Res 65:125–131. 152. Murakami S, Balmes G, McKinney S, Zhang Z, 168. Parr BA, McMahon AP (1995) Dorsalizing signal Givol D, de Crombrugghe B (2004) Constitutive Wnt-7a required for normal polarity of D-V and A-P activation of MEK1 in chondrocytes causes Stat1- axes of mouse limb. Nature 374:350–353. independent achondroplasia-like dwarfi sm and 169. Podbesek R, Edouard C, Meunier PJ, Parsons JA, rescues the Fgfr3-defi cient mouse phenotype. Genes Reeve J, Stevenson RW, Zanelli JM (1983) Effects of Dev 18:290–305. two treatment regimes with synthetic human para- 153. Nagata S (1997) Apoptosis by death factor. Cell thyroid hormone fragment on bone formation and 88:356–365. the tissue balance of trabecular bone in greyhounds. 154. Nakahara H, Bruder SP, Haynesworth SE, Holecek JJ, Endocrinology 112:1000–1006. Baber MA, Goldberg VM, Caplan AI (1990) Bone and 170. Praemer AP, Furner S, Rice DP (1992) Musculoskele- cartilage formation in diffusion chambers by subcul- tal Conditions in the United States. Park Ridge: tured cells derived from the periosteum. Bone American Academy of Orthopaedic Surgeons. 11:181–188. 171. Ramsauer M, D’Amore PA (2002) Getting Tie(2)d up 155. Nakajima A, Shimoji N, Shiomi K, Shimizu S, in angiogenesis. J Clin Invest 110:1615–1617. Moriya H, Einhorn TA, Yamazaki M (2002) Mecha- 172. Reddi AH (2001) Bone morphogenetic proteins: from nisms for the enhancement of fracture healing in rats basic science to clinical applications. J Bone Joint treated with intermittent low-dose human parathy- Surg Am 83A:S1–S6. roid hormone (1–34). J Bone Miner Res 17:2038– 173. Reimold AM, Grusby MJ, Kosaras B, Fries JW, Mori 2047. R, Maniwa S, Clauss IM, Collins T, Sidman RL, Glim- 156. Nakamura T, Hanada K, Tamura M, Shibanushi T, cher MJ, Glimcher LH (1996) Chondrodysplasia and Nigi H, Tagawa M, Fukumoto S, Matsumoto T (1995) neurological abnormalities in ATF-2-defi cient mice. Stimulation of endosteal bone formation by systemic Nature 379:262–265. injections of recombinant basic fi broblast growth 174. Roach HI, Erenpreisa J, Aigner T (1995) Osteogenic factor in rats. Endocrinology 136:1276–1284. differentiation of hypertrophic chondrocytes 157. Nakase T, Nomura S, Yoshikawa H, Hashimoto J, involves asymmetric cell division. J Cell Biol 131: Hirota S, Kitamura Y, Oikawa S, Ono K, Takaoka K 483–494. (1994) Transient and localized expression of bone 175. Roodman GD (2002) Role of the bone marrow micro- morphogenetic protein 4 messenger RNA during environment in multiple myeloma. J Bone Miner Res fracture healing. J Bone Miner Res 9:651–659. 17:1921–1925. 158. Nanes MS (2003) Tumor necrosis factor-alpha: 176. Rosen V, Thies RS (1992) The BMP proteins in molecular and cellular mechanisms in skeletal bone formation and repair. Trends Genet 8:97– pathology. Gene 321:1–15. 102. 44 Engineering of Functional Skeletal Tissues

177. Rouleau MF, Mitchell J, Goltzman D (1990) Charac- out mice. In: Durum S, Muegge K, eds. Cytokine terization of the major parathyroid hormone target Knockouts. Totowa: Humana Press, pp 89–101. cell in the endosteal metaphysis of rat long bones. J 193. Skripitz R, Andreassen TT, Aspenberg P (2000) Para- Bone Miner Res 5:1043–1053. thyroid hormone (1–34) increases the density of rat 178. Rousseau F, Saugier P, Le Merrer M, Munnich A, Del- cancellous bone in a bone chamber. A dose-response ezoide AL, Maroteaux P, Bonaventure J, Narcy F, study. J Bone Joint Surg Br 82:138–141. Sanak M (1995) Stop codon FGFR3 mutations in 194. St-Jacques B, Hammerschmidt M, McMahon AP thanatophoric dwarfi sm type 1. Nat Genet 10:11–12. (1999) Indian hedgehog signaling regulates prolifer- 179. Rowe NM, Mehara BJ, Luchs JS, Dudziak ME, Stein- ation and differentiation of chondrocytes and is brech DS, Illei PB, Fernandez GJ, Gittes GK, Lon- essential for bone formation. Genes Dev 13:2072– gaker MT (1999) Angiogenesis during mandibular 2086. distraction osteogenesis. Ann Plast Surg 42:470– 195. Street J, Bao M, deGuzman L, Bunting S, Peale FV Jr, 475. Ferrara N, Steinmetz H, Hoeffel J, Cleland JL, Daugh- 180. Ruther U, Garber C, Komitowski D, Muller R, Wagner erty A, van Bruggen N, Redmond HP, Carano RA, EF (1987) Deregulated c-fos expression interferes Filvaroff EH (2002) Vascular endothelial growth with normal bone development in transgenic mice. factor stimulates bone repair by promoting angio- Nature 325:412–416. genesis and bone turnover. Proc Natl Acad Sci USA 181. Ruther U, Komitowski D, Schubert FR, Wagner EF 99:9656–9661. (1989) C-fos expression induced bone tumors in 196. Sullivan R, Klagsbrun M (1985) Purifi cation of carti- transgenic mice. Oncogene 4:861–865. lage-derived growth factor by heparin affi nity chro- 182. Sahni M, Raz R, Coffi n JD, Levy D, Basilico C (2001) matography. J Biol Chem 260:2399–2403. STAT1 mediates the increased apoptosis and reduced 197. Suzuki A, Thies RS, Yamaji N, Song JJ, Wozney JM, chondrocyte proliferation in mice overexpressing Murakami K, Ueno N (1994) A truncated bone mor- FGF2. Development 128:2119–2129. phogenetic protein receptor affects dorsal-ventral 183. Sampath TK, Coughlin JE, Whetstone RM, Banach D, patterning in the early Xenopus embryo. Proc Natl Corbett C, Ridge RJ, Ozkaynak E, Oppermann H, Acad Sci USA 91:10255–10259. Rueger DC (1990) Bovine osteogenic protein is com- 198. Takada S, Stark KL, Shea MJ, Vassileva G, McMahon posed of dimers of OP-1 and BMP-2A, two members JA, McMahon AP (1994) Wnt-3a regulates somite and of the transforming growth factor-beta superfamily. tailbud formation in the mouse embryo. Genes Dev J Biol Chem 265:13198–13205. 8:174–189. 184. Sandborn WJ (2004) How future tumor necrosis 199. Tam CS, Heersche JN, Murray TM, Parsons JA (1982) factor antagonists and other compounds will meet Parathyroid hormone stimulates the bone apposition the remaining challenges in Crohn’s disease. Rev rate independently of its resorptive action: differen- Gastroenterol Disord 4:S25–S33. tial effects of intermittent and continuous adminis- 185. Scaffi di C, Fulda S, Srinivasan A, Friesen C, Li F, tration. Endocrinology 110:506–512. Tomaselli KJ, Debatin KM, Krammer PH, Peter ME 200. Tamai K, Zeng X, Liu C, Zhang X, Harada Y, Chang (1998) Two CD95 (APO-1/Fas) signaling pathways. Z, He X (2004) A mechanism for Wnt coreceptor acti- EMBO J 17:1675–1687. vation. Mol Cell 13:149–156. 186. Schipani E, Kruse K, Juppner H (1995) A constitu- 201. Thurston G, Gale NW (2004) Vascular endothelial tively active mutant PTH-PTHrP receptor in Jansen- growth factor and other signaling pathways in devel- type metaphyseal chondrodysplasia. Science 268:98– opmental and pathologic angiogenesis. Int J Hematol 100. 80:7–20. 187. Schultz DR, Harrington WJ Jr (2003) Apoptosis: pro- 202. Uemura A, Ogawa M, Hirashima M, Fujiwara T, grammed cell death at a molecular level. Semin Koyama S, Takagi H, Honda Y, Wiegand SJ, Yanco- Arthritis Rheum 32:345–369. poulos GD, Nishikawa S (2002) Recombinant angio- 188. Sekiya I, Larson BL, Smith JR, Pochampally R, Cui poietin-1 restores higher-order architecture of JG, Prockop DJ (2002) Expansion of human adult growing blood vessels in mice in the absence of stem cells from bone marrow stroma: conditions that mural cells. J Clin Invest 110:1619–1628. maximize the yields of early progenitors and evalu- 203. Urist MR (1965) Bone: formation by autoinduction. ate their quality. Stem Cells 20:530–541. Science 150:893–899. 189. Sekiya I, Vuoristo JT, Larson BL, Prockop DJ (2002) 204. Vaux DL, Flavell RA (2000) Apoptosis genes and In vitro cartilage formation by human adult stem autoimmunity. Curr Opin Immunol 12:719–724. cells from bone marrow stroma defi nes the sequence 205. Verma IM, Mulligan R, Beauset A (1988) Gene Trans- of cellular and molecular events during chondrogen- fer in Animals. UCLA Symposia on Molecular and esis. Proc Natl Acad Sci USA 99:4397–4402. Cellular Biology. New York: Alan R. Liss. 190. Sela-Donenfeld D, Kalcheim C (2002) Localized 206. Visnjic D, Kalajzic Z, Rowe D, Katavic V, Lorenzo J, BMP4-noggin interactions generate the dynamic Aguila HL (2004) Hematopoiesis is severely altered patterning of noggin expression in somites. Dev Biol in mice with an induced osteoblast defi ciency. Blood 246:311–328. 103:3258–3264. 191. Shiang R, Thompson LM, Zhu YZ, Church DM, 207. Von Bubnoff A, Cho K (2001) Intracellular BMP sig- Fielder TJ, Bocian M, Winokur ST, Wasmuth JJ (1994) naling regulation in vertebrates: pathway or network? Mutations in the transmembrane domain of FGFR3 Dev Biol 239:1–14. cause the most common genetic form of dwarfi sm, 208. Vortkamp A, Lee K, Lanske B, Segre GV, Kronenberg achondroplasia. Cell 78:335–342. HM, Tabin CJ (1996) Regulation of rate of cartilage 192. Shinbrot E, Moore M (1998) Cooperation between the differentiation by Indian hedgehog and PTH-related TNF receptors demonstrated by TNF receptor knock- protein. Science 273:613–622. Osteogenic Growth Factors and Cytokines and Their Role in Bone Repair 45

209. Vortkamp A, Pathi S, Peretti GM, Caruso EM, 220. Yamashita H, Ten Dijke P, Heldin CH, Miyazono K Zaleske DJ, Tabin CJ (1998) Recapitulation of signals (1996) Bone morphogenetic protein receptors. Bone regulating embryonic bone formation during post- 19:569–574. natal growth and in fracture repair. Mech Dev 221. Yasko AW, Lane JM, Fellinger EJ, Rosen V, Wozney 71:65–76. JM, Wang EA (1992) The healing of segmental bone 210. Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA, defects induced by recombinant human bone mor- Hanahan D, Shapiro SD, Senior RM, Werb Z (1998) phogenetic protein (rhBMP-2). J Bone Joint Surg MMP-9/gelatinase B is a key regulator of growth 74A:659–671. plate angiogenesis and apoptosis of hypertrophic 222. Yeh LC, Lee JC (1999) Osteogenic protein-1 increases chondrocytes. Cell 93:411–422. gene expression of vascular endothelial growth 211. Wallach D, Varfolomeev EE, Malinin NL, Goltsev YV, factor in primary cultures of fetal rat calvaria cells. Kovalenko AV, Boldin MP (1999) Tumor necrosis Mol Cell Endocrinol 153:113–124. factor receptor and fas signaling mechanisms. Annu 223. Ying Y, Zhao GQ (2001) Cooperation of endoderm- Rev Immunol 17:331–367. derived BMP2 and extraembryonic ectoderm- 212. Wang EA, Rosen V, Cordes P, Hewick RM, Kriz MJ, derived BMP4 in primordial germ cell generation in Luxenberg DP, Sibley BS, Wozney JM (1988) Purifi ca- the mouse. Dev Biol 232:484–492. tion and characterization of other distinct bone- 224. Young-In-Lee F, Choi YW, Behrens FF, DeFouw DO, inducing factors. Proc Natl Acad Sci USA 85:9484– Einhorn TA (1998) Programmed removal of chon- 9488. drocytes during endochondral fracture healing. J 213. Wang Z-Q, Ovitt C, Grigoriadis AE, Mohle-Steinlein Orthop Res 6:144–149. U, Ruther U, Wagner EF (1992) Bone and haemato- 225. Yu J, Tian S, Metheny-Barlow L, Chew LJ, Hayes AJ, poietic defects in mice lacking c-fos. Nature 360: Pan H, Yu GL, Li LY (2001) Modulation of 741–745. endothelial cell growth arrest and apoptosis by vas- 214. Westendorf JJ, Kahler RA, Schroeder TM (2004) Wnt cular endothelial growth inhibitor. Circ Res 89: signaling in osteoblasts and bone diseases. Gene 1161–1167. 341:19–39. 226. Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai 215. Wilkie AO, Slaney SF, Oldridge M, Poole MD, Ash- N, Soker S, Olsen BR (2001) Tissue specifi c regulation worth GJ, Hockley AD, Hayward RD, David DJ, of VEGF expression during bone development Pulleyn LJ, Rutland P, Malcolm S, Winter RM, requires Cbfa1/Runx2. Mech Dev 106:97–106. Reardon W(1995) Apert syndrome results from local- 227. Zhai Y, Ni J, Jiang GW, Lu J, Xing L, Lincoln C, Carter ized mutations of FGFR2 and is allelic with Crouzon KC, Janat F, Kozak D, Xu S, Rojas L, Aggarwal BB, syndrome. Nat Genet 9:165–172. Ruben S, Li LY, Gentz R, Yu GL (1999) VEGI, a novel 216. Wodarz A, Nusse R (1998) Mechanisms of Wnt sig- cytokine of the tumor necrosis factor family, is an naling in development. Annu Rev Cell Dev Biol 14: angiogenesis inhibitor that suppresses the growth of 59–88. colon carcinomas in vivo. FASEB J 13:181–189. 217. Wozney JM (1989) Bone morphogenetic proteins. 228. Zhang H, Bradley A (1996) Mice defi cient for BMP2 Prog Growth Factor Res 1:267–280. are nonviable and have defects in amnion/chorion 218. Xiao Z, Latek R, Lodish HF (2003) An extended and cardiac development. Development 122:2977– bipartite nuclear localization signal in Smad4 is 2986. required for its nuclear import and transcriptional 229. Zhou T, Edwards CK 3rd, Yang P, Wang Z, Blueth- activity. Oncogene 22:1057–1069. mann H, Mountz JD (1996) Greatly accelerated 219. Xu X, Weinstein M, Li C, Deng C (1999) Fibroblast lymphadenopathy and autoimmune disease in lpr growth factor receptors (FGFRs) and their roles in mice lacking tumor necrosis factor receptor I. J limb development. Cell Tissue Res 296:33–43. Immunol 156:2661–2665.