Abstract
Bone remodeling is essential for adult bone homeostasis. It comprises two phases: bone formation and resorption. The balance between the two phases is crucial for sustaining bone mass and systemic mineral homeostasis. This review highlights recent work on physiological bone remodeling and discusses our knowledge of how systemic and growth factors regulate this process.
The skeleton is an extremely specialized and dynamic organ that undergoes continuous regeneration. The process of bone modeling is responsible for the formation and maintenance of the shape of bone. Bone modeling leads to the acquisition of peak bone mass. It begins in fetal life and continues until skeletal maturity (i.e., epiphyseal plate closure or completion of longitudinal growth of the skeleton). Bone modeling occurs by removal of bone from one site and formation of bone at different sites. The bone regeneration process persists even after skeletal maturity by periodic replacement of old bone with newly formed bone at the same location, called remodeling. Bone remodeling is required for repair of old damaged bone due to daily physical load and prevention of aging effects and its consequences. Impairment in the bone remodeling process often results in the progression to osteoporosis, a major worldwide health concern. The overall process of bone remodeling is a tightly controlled and coordinated process regulated by a number of cell types. The maintenance of physiological bone remodeling and systemic mineral homeostasis requires balance between bone formation and bone resorption (121, 150). In this review, we discuss the recent observations of the role of systemic hormones and growth factor signals in regulation of bone remodeling.
Bone Remodeling
To accomplish normal physiological bone remodeling, the proper coupling of bone formation and bone resorption requires direct communication among different bone cells. Cells of the osteoblast lineage (osteoblasts, osteocytes, and bone-lining cells) and bone-resorbing cells (osteoclasts), together with their precursor cells, are organized in specialized units called bone multicellular units (BMU) (33). Osteoblasts originate from mesenchymal stem cells in the bone marrow stroma and are responsible for bone matrix synthesis and its subsequent mineralization. On the other hand, osteoclasts are large, multinucleated giant cells formed from the fusion of mononuclear progenitors of the monocyte/macrophage in a process termed osteoclastogenesis. The primary purpose of the BMU is to facilitate normal physiological bone remodeling.
There are many factors that can be involved in regulating the precise bone remodeling cycle (Table 1). The remodeling cycle is composed of seven sequential phases, namely, quiescence, activation, resorption, reversal, formation, mineralization, and termination. Activation precedes resorption, which precedes reversal, with termination as the last step (FIGURE 1).
Table 1.
Hormones and factors involved in BMU remodeling sequence
| Remodeling Phases | Hormones and Factors Regulating Bone Remodeling |
|---|---|
| Activation phase | +, PTH, IGF-1, IL-1, IL-6, PGE2, Calcitriol, TNF-α |
| −, Estrogen | |
| Osteoclast recruitment and resorption phase | +, RANKL, M-CSF, αvβ3 integrins, IL-1β, IL-1α, TNF-α, retinoic acid, S1P |
| −, OPG, GM-CSF, estrogen, Calcitonin, IL-4, IL-18, TGF-β | |
| Reversal phase | TGF-β, IGF-1, IGF-2, BMPs, PDGF, or FGF |
| Osteoblast recruitment and formation phase | +, WNTs, BMPs, IGF-1, FGF-2, FGF-18, PDGFs, PTH, 1,25(OH)2 vitamin D3, Runx2, TGF-β, CT-1, ephrin B2 |
| −, PDGFs, glucocorticoids, leptin, pyrophosphate, Sema4D | |
| Termination phase | Feedback signal of normal sclerostin expression |
S1P, sphingosine-1-phosphate; CT-1, cardiotrophin-1; Sema4D, semaphorin 4D.
FIGURE 1.
Physiological bone remodeling
Bone remodeling follows coordination of distinct and sequential phases of the process. The remodeling cycle is composed of six sequential phases, namely, quiescence, activation, resorption, reversal, formation, and termination. Activation precedes resorption, which precedes reversal, with mineralization as the last step. The first stage of bone remodeling involves detection of an initiating remodeling signal, which has usually been described as resorption by osteoclasts. In the resorption phase, osteoblasts respond to signals generated by osteocytes or direct endocrine activation signals, recruiting osteoclast precursors to the remodeling site. The resorption phase is of limited duration depending on level of the stimuli responsible for osteoclast differentiation and activity. It is followed by the reversal phase that is characterized by disappearance of almost all osteoclasts. The formation phase is distinct by complete replacement of osteoclastic cells with osteoblastic cells. The termination signals of bone remodeling include the terminal differentiation of the osteoblast. The resting bone surface environment is maintained until the next wave of remodeling is initiated.
Systemic Regulation of Bone Remodeling
The bone remodeling process is controlled by various local and systemic factors, and their expression and release, in a well organized manner. Calcitonin (CT), parathyroid hormone (PTH), vitamin D3 [1,25(OH)2 vitamin D3] and estrogen are the major hormonal regulators of osteoclastic bone resorption. Secretion of the first three is driven by the requirement to control the physiological serum calcium level. In addition to systemic hormonal regulation, it has become more and more apparent that growth factors such as IGFs, TGF-β, FGFs, EGF, WNTs, and BMPs play significant roles in regulation of physiological bone remodeling (FIGURE 2). Because of our accumulated and recent knowledge of the critical function of these growth factors in the skeleton, we have been able to turn from classical treatment of skeletal diseases with hormones to manipulation of some of the growth factor pathways to treat diseases such as osteoporosis.
FIGURE 2.

Systemic and growth factor regulation of bone remodeling
PTH induces differentiation of committed osteoblast precursors, induces RUNX2 expression in osteoblasts, increases osteoblast numbers, and extends osteoblast survival. PTH stimulates the proliferation and differentiation of osteoprogenitors to mature osteoblasts via IGF-1. Along with IGF-I, PTH induces RANKL and MCSF from mature osteoblasts to promote osteoclastogenesis. PTH elevates cAMP levels and inhibits Mef2-stimulated Sost promoter activity, leading to decreased expression of sclerostin and an elevated bone formation rate. 1,25(OH)2D3 stimulates osteoblastogenesis via differentiation of mesenchymal stem cells (MSCs) to osteoblasts. Calcitonin increases osteoblast proliferation and suppresses bone resorption by inhibiting the activity of osteoclasts. Estrogen inhibits bone resorption by directly inducing apoptosis of the bone-resorbing osteoclasts. Androgens can also indirectly inhibit osteoclast activity and bone resorption via effects on osteoblasts/osteocytes and the RANKL/RANK/OPG system. Besides systemic hormonal regulation, other growth factors, such as IGFs, TGF-β, FGFs, EGF, WNTs, and BMPs, also play a significant role in regulation of physiological bone remodeling.
PTH and PTHrP
PTH is a hormone synthesized and secreted by the parathyroid glands. The main function of PTH is to maintain blood calcium homeostasis. In addition, PTH regulates bone mass in an endocrine manner (52). PTH-related peptide (PTHrP) is a genetically related peptide that can mimic several functions of PTH. While PTH and PTHrP share nearly 70% of homology within the first 13 residues of their amino-terminal domain, the domain essential for their bioactivity (47, 99), cross talk between COOH and NH2 domains of PTHrP have been reported to trigger signal transduction in mesenchymal stem cells and osteoblast-like cells (23, 60, 177). In contrast to PTH, PTHrP is expressed in almost all tissues and exerts its effect in an autocrine/paracrine manner. PTH1R is the common receptor for both PTH and PTHrP, and is expressed in bone and cartilage cells of the mesenchymal lineage (72, 112, 193). PTHrP regulates the proliferation and differentiation of growth plate chondrocytes, and PTHrP-null mice are characterized by the shortening of the growth plate due to accelerated chondrocyte maturation (100). Targeted disruption of PTHrP in embryonic mice is lethal and causes impaired endochondral bone development (74, 96). PTH1R-null mice exhibit a similar skeletal phenotype. Apart from articular chondrocytes, PTHrP is also expressed by early osteoblastic lineage cells, suggesting a role for PTHrP in regulation of cells in bone (75). PTHrP-overexpressing mice have shorter and thicker limbs. Furthermore, both PTHrP heterozygous-null mice and osteoblast-specific PTHrP knockout mice have decreased bone volume and defective bone, suggesting the anabolic role of PTHrP in bone formation and leading to testing PTHrP analogs for treatment of osteoporosis (103, 124).
PTH can exert both catabolic and anabolic effects on bone (37). It is well established that daily injections of low doses of PTH increase bone mass in animals and humans (36, 127). PTH has no direct effect on osteoclasts. Continuous administration of PTH or PTHrP induces bone resorption indirectly through their actions on osteoblastic cells (151, 176). Several effects of PTH on induction of osteoclast formation are mediated through the osteoblast by stimulation of RANKL and inhibition of OPG mRNA expression (105). It is well recognized that constant high levels of PTH facilitate bone resorption, whereas low and intermittent doses of PTH cause an increase in bone mineral density at the lumbar spine and hip, and promote new bone. PTH's anabolic actions are thought to be via its effects to increase the proliferation and differentiation of osteoblasts in vitro and in vivo (43, 90, 137, 163, 172), decrease osteoblast apoptosis (70, 71, 116), and activate bone-lining cells (38, 102).
A number of investigations into the mechanisms of PTH's anabolic effects have elucidated the role of several growth factor and chemokine pathways. For instance, PTH induces the synthesis of IGF-1 to stimulate osteoblast proliferation and differentiation as well as indirectly stimulate osteoclast activity (17, 111, 181). It was recently shown that PTH-mediated new bone formation is attenuated by Dkk1, an inhibitor of Wnt signaling showing the role of the Wnt pathway in PTH action (57). In addition, activation of the PTH receptor in T lymphocytes has a role in PTH-induced bone formation by increasing Wnt10b synthesis by these cells (11). Mice expressing a constitutively active PTH/PTH-related peptide (PTHrP) receptor (PPR) in osteocytes exhibit increased bone mass and bone remodeling, which is sufficient for inhibiting SOST expression (another Wnt pathway inhibitor) through LRP5-dependent and -independent mechanisms (12, 141, 158). These are some of the many studies showing cross talk between PTH and Wnt signaling is an important mechanism regulating bone formation. Finally, PTH-induced osteoblastic expression of MCP-1 leads to recruitment and differentiation of osteoclast precursors and is required for enhanced bone formation (106, 174). These data show a role for MCP-1 in the anabolic effects of PTH. Thus the complexity and knowledge of PTH and PTHrP regulation of the skeleton has opened up opportunities for new treatment modalities.
1,25(OH)2 Vitamin D3
Vitamin D is essential for normal development and maintenance of the skeleton. Active 1α,25(OH)2D3 plays a central role in calcium and bone homeostasis through binding to the vitamin D receptor (VDR) present mainly in intestine, bone, kidney, and parathyroid gland. VDR knockout mice are phenotypically very similar to those with vitamin D deficiency (108, 194). In contrast to its classical endocrine role, there is accumulating evidence for an autocrine role for vitamin D in bone (5, 6). The enzyme CYP27B1, which converts 25D to the active form 1,25D, promotes osteoblast proliferation and maturation in human osteoblasts in vitro. It has been shown that the expression of CYP27B1 is dependent on the stage of osteoblast maturation, and expression in vitro is maximal immediately before mineralization reaches its peak (7). Gene deletion of CYP27B1 in mice caused osteomalacic abnormalities thought to be due largely to hypocalcaemia and hyperparathyroidism as a result lead to cortical porosity (149). In addition, recently it has been shown that 25D metabolism in osteoclast lineage cells appears to promote osteoclast differentiation, ameliorate osteoclast activity, and promote coupling of bone resorption to formation (86, 87). In contrast, chronic 1,25(OH)2D3 treatment causes the OPG gene to become insensitive to repression, thereby facilitating the anabolic phase of the vitamin D effect on bone (89).
It has been recently shown that 1,25(OH)2D3 treatment enhanced mineralization of the osteoblast and expression of osteocyte markers (dentin matrix protein-1 and FGF-23) in induced pluripotent stem (iPS)-derived osteoprogenitors (iPSop cells), suggesting that 1,25(OH)2D3 is a potent promoter of the osteoblast-osteocyte transition (77). In support of this notion, there is also evidence for presence of VDR transcripts in osteocytes and direct activation of 25(OH)D3 in osteocytes (34, 133).
In summary, 1,25(OH)2 vitamin D3 appears to have both endocrine and autocrine actions in bone.
Calcitonin
Calcitonin (CT) is a 32-amino acid hormone that is secreted by the thyroid C cells. Tissue-specific alternative splicing of the calcitonin gene results in the production of another peptide, calcitonin gene-related peptide-1 (CGRP-1). Most of the effects of both peptides are exerted by binding to the calcitonin receptor (CTR) and calcitonin receptor-like receptor, members of the family of GPCRs that are coupled to both PKA and PKC via Gs and Gq, respectively (44, 67, 109). CT mediates its actions through the CTR expressed on osteoclasts (65). Osteoclast-specific deletion of the CTR increased bone formation by inhibiting the release of sphingosine 1-phosphate (S1P) from osteoclasts (82). CT inhibits both basal and stimulated resorption via directly causing a loss of the ruffled border of osteoclasts and reducing osteoclast numbers over time (27, 28).
Although calcitonin receptors are thought not to be present on osteoblast-like cells, CT is reported to interact with osteoblasts in some systems. CT has been reported to enhance osteoinduction by rhBMP-2 on osteoblasts (144). CT also increases the extracellular level of insulin-like growth factors (IGF-1 and IGF-2) in cultures of human osteoblast-like cells (42). In addition, CT may also prevent osteoblast and osteocyte apoptosis (155). Despite these reports, any effect of CT on osteoblasts remains unclear. Evidence suggests that osteocytes can also express the CTR (54, 148), and its expression declines with age (53). CT mainly acts to complement the function of PTH by counteracting the increased bone resorption caused by PTH (26). Yet, these recent observations may change our view of CT.
Estrogen
Estrogen is the one of the major hormonal regulators of bone metabolism in both women and men. Estrogens attenuate osteoclastogenesis and stimulate osteoclast apoptosis. However, its regulation of bone metabolism is complex. Female mice in which the ERα was specifically deleted in mature osteoclasts using the cathepsin K promoter exhibited decreased bone volume due to an increase in the number of osteoclasts, whereas deletion of ERα in male mice does not alter bone mass. Female mice with ERα deletion in osteoblasts had decreased cancellous (proximal tibia, vertebra, and distal femur) and cortical (tibial midshaft and L5 vertebral cortex) bone mass due to a reduction in osteoblast activity rather than osteoclast number, indicating that ERα in osteoblasts is required for appropriate bone mass (123). Mice with deletion of ERα in mesenchymal progenitors have decreased proliferation and differentiation of periosteal cells due to decreased canonical Wnt signaling, which results in low cortical bone mass (3).
In osteoclasts, estrogen blocks RANKL/M-CSF-induced activator protein-1-dependent transcription through a reduction of c-Jun activity, thus suppressing RANKL-induced osteoclast differentiation (165). In addition, estrogen has also been shown to modulate the production of a number of bone-resorbing cytokines, including IL-1, IL-6, TNF-α, M-CSF, and prostaglandins (4, 85, 117, 145). T cells are an essential mediator of the bone-wasting effects of estrogen deficiency in vivo, and estrogen prevents osteoclastic bone resorption through suppressing the production of TNF-α from T cells (25). Furthermore, estrogen has been shown to inhibit osteoblast apoptosis and to increase osteoblast lifespan via activation of the Src/Shc/ERK signaling pathway (91) and downregulation of JNK (92).
Taken together, estrogen modulates both osteoclast and osteoblast activities, which subsequently lead to inhibition of bone remodeling, decreased bone resorption, and maintenance of bone formation. Estrogens regulate cortical and trabecular bone mass via osteoblast progenitors and osteoclasts, respectively (3, 118).
Androgens
Estrogen is a well known and intensely studied hormonal regulator of postmenopausal bone health in women. However, androgens also have several beneficial effects on development and maintenance of bone mass in both men and women. Androgen deficiency results in increased bone remodeling and bone loss in men (79). However, some part of the bone loss is due to reduced levels of estrogen, which is derived from aromatization of testosterone (41). Androgens can modulate growth plate maturation and closure, and thus affect longitudinal bone growth. In addition, androgens regulate trabecular and cortical bone mass, and inhibit bone loss (184).
Global deletion of the AR in male mice resulted in high bone turnover, increased resorption, decreased trabecular and cortical bone volume, as well as decreased periosteal bone formation (80). In contrast, mice with conditional deletion of the AR in osteoblasts and osteocytes have normal cortical bone, showing that androgens modulate periosteal bone formation without direct involvement of the mature osteoblast lineage (31, 140, 169). Probably, these effects might result from improvement in physical activity due to stimulatory effects of androgen on muscle mass and strength, which leads to activation of bone-forming sites and stimulation of osteoprogenitors. In contrast, these mice show decreased trabecular bone mass, indicating a role for the AR in trabecular osteoblastic cells. The mechanisms responsible for this positive effect of the AR on trabecular bone mass remain elusive (31, 140, 169).
Recent evidence suggests that androgens can indirectly inhibit osteoclast activity and bone resorption via effects on osteoblasts/osteocytes and the RANKL/RANK/OPG system (46). Androgen increases collagen, type 1 protein and mRNA levels, osteocalcin secretion, and alkaline phosphatase activity in transformed clonal human osteoblastic cells, which leads to their increased differentiation and mineralization (15, 76). As well, suppression of either testosterone or estradiol increases the PTH bone-resorbing effects in men (104), showing the interplay between the sex steroids and the parathyroid axis. In conclusion, androgens directly and indirectly modulate osteoblast and osteoclast activities, respectively, and may maintain bone mass through both the AR and ERα. Further studies are needed to delineate the role of androgens on the skeleton in each sex.
Thyroid Hormone
The hypothalamic-pituitary-thyroid axis plays a major role in skeletal development, peak bone mass achievement, and regulation of bone turnover (51). Hypothyroidism causes reduction in osteoblast formation and osteoclast resorption, and results in low bone turnover or slowing of the bone remodeling process (9, 134). In contrast, in thyrotoxicosis, there is an increase in osteoblast and osteoclast activity, and an increase in bone turnover with an impaired bone formation cycle, resulting in a remodeling process favoring rapid resorption (45). In adults, hyperthyroidism is associated with increased bone remodeling, reduced bone mineral density (BMD), and increased fracture risk, mainly in postmenopausal women (128, 135).
In vitro, the biologically active derivative of thyroxine, triiodothyronine (T3), has effects on both osteoblasts and osteoclasts. T3 has dual actions on osteoblasts: it has been reported that T3 promotes osteoblast differentiation and inhibits osteoblast proliferation in both osteoblastic cell lines and primary calvarial osteoblasts. In osteoclasts, T3 stimulates osteoclast activity either directly (73) or indirectly through cytokines (19).
The action of T3 requires thyroid hormone nuclear receptors (TRs). It has been reported that both TRα and TRβ receptors play a role in mediating TH actions on bone remodeling (18, 128, 131). TRα-deficient mice acquire a higher bone mass than wild-type mice, whereas pharmacological activation of TRβ with GC-1 (a TRβ agonist) spares the skeleton from bone loss, yielding the conclusion that T3 mediates its effects in bone via TRα (10, 45). Overall, like the other members of the nuclear receptor superfamily, thyroid hormones also have effects on both osteoblast and osteoclast activities, and are essential for bone mineral homeostasis, normal skeletal growth, and maintenance of bone mass.
Glucocorticoids
Glucocorticoids (GCs) can affect physiological bone remodeling via accelerating bone resorption and reducing bone formation (68, 183). Prolonged glucocorticoid therapy or excessive exposure (Cushing's syndrome) decreases bone mineral density (BMD) and may lead to development of glucocorticoid-induced osteoporosis (GIO) (113, 114). Bone deterioration is more apparent in trabecular than in cortical bone in GIO and depends on dose and duration of glucocorticoid exposure (21). In adult bone, functional glucocorticoid receptors (GR) are mainly found in pre-osteoblast/stromal cells and osteoblasts (1). Consistent with these observations, GCs fail to decrease bone formation in osteoblast-specific (Runx2-Cre) GR knockout mice, suggesting that negative effects of GCs on bone formation are mediated through osteoblastic GR (157). Similarly, GCs stimulate osteoclast numbers by suppressing synthesis of OPG and stimulating RANKL synthesis by pre-osteoblast/stromal cells, with the decrease in the OPG-to-RANKL ratio supporting osteoclast differentiation and net bone resorption (61, 84). It has been reported that long-term use of GCs reduces bone formation either via direct inhibition of osteoblast proliferation and differentiation (suppression of Runx2 and type I collagen) or by the augmentation of apoptosis rates of mature osteoblasts and osteocytes (153, 154, 183). In addition, GCs negatively regulate secretion of androgens and estrogens by inhibiting gonadotropin secretion and finally increase bone resorption (152). GCs also induce negative calcium balance by decreasing intestinal calcium absorption and increasing urinary calcium excretion (49). Taken together, direct GC-GR signaling in bone can reduce bone mass as well as through indirectly affecting systemic regulators of bone remodeling.
Growth Hormone
Growth hormone (GH) is a peptide hormone secreted from the pituitary gland under the control of the hypothalamus. It has been reported that both GH-deficient humans and GHR−/− mice have reduced longitudinal bone growth (159, 171). GH supplementation to GH-deficient adults and children has positive skeletal effects, including increases in BMD and bone turnover markers (98). GH, along with its binding protein (GHBP), regulates growth directly through the GH receptor (GHR) and indirectly by stimulating liver and skeletal IGF-1 expression (143). It has been reported that GH stimulates osteoblast proliferation and collagen production either directly (132) and/or indirectly by increasing IGF-1 and IGF binding protein (IGFBP) production (40, 190, 191). Consistent with this, hIGF-1 treatment of GHR−/− mice restored normal bone formation (179). GH also stimulates bone resorption, although it is not clear whether these effects are results of direct GH stimulation or mediated by osteoblastic IGF-1 or IGFBP (138, 168). Finally, administration of GH along with PTH in rats increases bone growth and bone formation, decreases bone resorption, and has a synergistic effect on increasing bone density and bone mass (56). Recent work has shown that GH has direct effects on the osteocyte, further adding to the complexity of this hormone's actions (110).
Growth Factor Regulation of Bone Remodeling
Bone Morphogenetic Proteins
Bone morphogenetic proteins (BMPs) are members of the TGF-β superfamily and regulate differentiation of bone marrow mesenchymal cells into components of bone, cartilage, or adipose tissue. It has been reported that BMP-2, -4, -5, -6, and -7 all have strong osteogenic capability and that their signaling is essential for bone remodeling and maintenance of bone mass through activation of BMP type 1A and type 1B receptors (97, 125). BMP-mediated signaling increases the expression of Runx2 and Osx, mainly through the Smad and MAPK pathways.
One of the highly studied BMP family members, BMP-2, enormously increases osteocalcin expression, and short-term expression of BMP-2 is sufficient to induce bone formation (64, 139). It has been reported that loss of both BMP-2 and BMP-4 resulted in severe impairment of osteogenesis (8). In addition, BMP-7 upregulated ALP activity and increased mineralization, resulting in increased osteoblast differentiation (164). BMP-2 has osteoinductive properties and improves healing of open tibial fractures (20). BMP-2 has been used extensively in orthopedic procedures, but some of the trials have recently been criticized (20, 22, 55).
Transforming Growth Factor-β
The transforming growth factor (TGF)-β signaling pathway is known to control bone remodeling and maintenance. Compared with wild-type mice, TGF-β1 knockout mice have decreased bone mineral content in the proximal tibial metaphysis and shorter tibiae (48). In contrast, transgenic mice overexpressing TGF-β2 develop osteoporosis in association with increased osteoclastic bone resorption (39). Paradoxically, TGF-β1 stimulates apoptosis of osteoclasts (63). Thus the role of TGF-β appears complex and to involve both the osteoclast and the osteoblast.
TGF-β has dual effects on osteoblasts. TGF-β has been shown to stimulate expression of early osteoblast differentiation and bone matrix proteins by osteoblasts. However, TGF-β inhibits late osteoblast differentiation. TGF-β binds to its receptors (TβRI and II) and activates Smad2 and Smad3 via phosphorylation. Furthermore, phosphorylated Smad3 physically complexes with Runx2. This reduces the transcriptional and osteogenic activity of Runx2 through the involvement of histone deacetylases (HDAC4 and HDAC5) (2).
TGF-β and BMPs show opposite roles in osteoblast differentiation by using different Smad molecules. Endogenous TGF-β opposes BMP signaling by inducing the inhibitory Smads and subsequently inhibits late-stage osteoblast maturation (30). TGF-β also signals with other bone remodeling systems. TGF-β and PTH signaling coordinate each other in regulation of osteoblastogenesis and bone remodeling signaling (156). The TβRII forms an endocytic complex with the PTH1R and phosphorylates the PTH1R cytoplasmic domain, accelerating downregulation of PTH1R signaling (156). In support of this, deletion of TβRII in osteoblasts in mice results in a bone phenotype similar to hyperactivation of the PTH1R (156). TGF-β proteins are present in their latent form in the bone matrix, and osteoclasts are able to release and activate TGF-β from the bone matrix via osteoclastic bone resorption (175). As a result, active TGF-β1 recruits bone mesenchymal stem cells to the bone remodeling area, via phosphorylation of Smad2 and Smad3, leading to coupling of bone resorption with bone formation (175).
Epidermal Growth Factors and Receptor
The epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein. EGFR deficiency leads to early lethality with a few surviving Egfr-null pups displaying craniofacial alterations, delayed primary ossification of the cartilage, and impairment in trabecular bone formation (180). It was recently found that decreasing EGFR expression in pre-osteoblasts and osteoblasts in mice results in reduced osteoblastogenesis and increased bone resorption, finally leading to decreased trabecular and cortical bone (196). Thus osteoblastic EGFR signaling plays an anabolic role in bone metabolism (198). However, all EGF-like ligands inhibit osteoblast differentiation (198). Moreover, they suppress gene expression of osteoblast-specific transcription factors Runx2 and osterix, and osteoblastic markers such as alkaline phosphatase, bone sialoprotein (BSP), and osteocalcin (198). It has been shown that attenuation of EGFR activity in osteoblast lineage cells decreases the number of osteoprogenitor cells, showing that EGFR stimulates osteoprogenitor proliferation and that EGFR signaling is required to maintain the osteoprogenitor population (29, 198). Apart from these actions, EGF and TGF-α stimulate bone resorption in fetal rat long bones and human marrow, suggesting that EGFR signaling also regulates osteoclastogenesis and bone resorption. It has been reported, using osteoblast/osteoclast co-culture, that this occurs by decreasing osteoblastic expression of OPG and by increasing osteoblastic expression of MCP-1 but having no effect on RANKL expression (197).
Fibroblast Growth Factors
Fibroblast growth factors (FGF) belong to a family of heparin-binding growth factors. Basic FGF (FGF-2) is expressed by osteoblastic cells and is an important modulator of cartilage and bone growth and differentiation. Ablation of endogenous FGF-2 in mice decreases trabecular bone and bone formation rates (130). FGF-2 also stimulates the differentiation of mesenchymal bone marrow stromal cells to adipocytes or osteoblasts (120, 186). Endogenous FGF-2 was found to be necessary for the bone anabolic effects of PTH and BMP-2 in mice (136, 161). Another FGF family member, FGF-18, is an essential regulator of the osteogenic differentiation of murine mesenchymal stem cells (58), and FGF-18-deficient mice display delayed ossification, showing an important role of FGF-18 in osteogenesis (142). In contrast, FGFR1 inactivation leads to increased bone mass, indicating that signaling through this receptor negatively controls osteoblast maturation and bone formation in vivo (69). FGFR2 plays a role in bone formation, and deletion of FGFR2 results in atypical function of osteoblasts, decreased proliferation of osteoprogenitor cells, and decreased bone mineral density. Wnt and FGFR signaling act together to control mesenchymal stem cell differentiation, suggesting that cross talk between FGF/FGFR signaling and Wnt-β-catenin signaling regulate osteoblast function (122).
FGF-23 is an important member of the FGF family produced primarily by osteocytes and by osteoblasts that acts on the kidney and the parathyroid gland (119, 162), and has a major role in phosphate homeostasis and signaling between bone and the kidney. The production of FGF-23 by osteocytes is also regulated by systemic hormones, mainly PTH and 1,25(OH)2 vitamin D3 (66, 81, 88, 167). FGF-23 negatively regulates erythropoiesis in peripheral blood and bone marrow in young adult mice (32). Recently, it has been shown that FGF-23 has a direct effect on the differentiation of bone marrow stromal cells (107). The effects of FGF-23 on bone mineralization are thought to be exerted through interrelated physiological pathways associated with phosphate homeostasis (166). However, it has been shown that excess FGF-23 can negatively regulate bone mineralization, and this bone mineralization effect of FGF-23 is independent of systemic phosphate levels (170). We still do not know all of the roles and actions of FGF-23, and since it is the first marker to increase in chronic kidney disease (CKD), the gap in knowledge of this growth factor is an impediment to treatment of CKD (160, 178).
Insulin-like Growth Factor-1
Insulin-like growth factor-1 (IGF-1) is the most abundant growth factor deposited in the bone matrix and stimulates cell proliferation and function, and survival of osteoblasts. The presence of IGF-1 in osteoblasts and osteoclasts suggests its involvement in active bone growth and remodeling (101). The primary function of IGF-1 in the bone matrix is to maintain bone mass and skeletal homeostasis during bone remodeling (185). Mice lacking functional global IGF-1 exhibit severe deficiency in bone formation and a 60% deficit in peak bone mineral density (BMD) (16). Like the TGF-βs, IGF-1 has been implicated in the coupling process through its actions on MSC differentiation (59, 126, 195). Furthermore, IGF-1 promotes osteoclast differentiation, and IGF-1-null mice show high bone mass and impaired bone resorption, suggesting a positive role for IGF-1 in regulation of osteoclast differentiation (182). IGF-1 regulates the expression of RANKL and RANK, and facilitates normal physiological interaction between the osteoblast and the osteoclast (182). Liver is the primary source of serum IGF-1, and serum IGF-1 levels are tightly controlled by pituitary growth hormone (GH), whereas numerous other tissue factors along with GH regulate tissue IGF-1. There are several reports demonstrating the synergistic effects of IGF-1 and PTH on bone remodeling and establishing the involvement of locally produced IGF-1 in the anabolic effects of PTH (188, 189, 192).
WNT and WNT Antagonists
WNT signaling is implicated in proximal-distal outgrowth and dorsoventral limb pattern formation during skeletal development. Genetic studies in human and animal models suggest that the canonical Wnt/β-catenin pathway, together with BMP signaling and Runx2, has a significant vital role in osteoblast differentiation, skeletal development, and bone formation (95, 129). WNT signaling represses mesenchymal stem cell commitment to the chondrogenic and adipogenic lineages and enhances differentiation of osteoblastic lineages (35, 78, 83). Osteoblast and osteocyte WNT-β-catenin signaling has a negative effect on osteoclast differentiation and bone resorption through the increased secretion of osteoprotegerin (50, 62, 93).
Canonical WNT signal transduction involves stabilization of β-catenin by inhibiting GSK-3-β-mediated β-catenin phosphorylation. On the other hand, activation of noncanonical pathways by osteoblast-expressed WNT5a stimulates differentiation of osteoclast precursors and osteoclastogenesis (115). WNT 10b−/− mice had decreased trabecular bone and serum markers (13). WNT10b is the best-characterized bone formation-promoting canonical WNT and can trigger osteoblast Wnt/β-catenin signaling in a paracrine manner (13, 14, 173). It has been reported that WNT6a and WNT10a modulate the fate of mesenchymal cell differentiation to osteoblasts or adipocytes, and thereby control physiological bone remodeling (24). Taken together, WNT signaling has a direct and indirect effect on osteoblast and osteoclast bone cell lineages, leading to an overall increase in bone formation.
Dickkopf family members (Dkk1 and Dkk2) and secreted frizzled-related proteins (Sfrps) are families of extracellular proteins that negatively modulate canonical Wnt signaling (93). Another of these is sclerostin, a secreted glycoprotein encoded by the SOST gene, which binds to LRP5/6 receptors and inhibits BMP- and WNT-stimulated bone formation (94). Neutralizing monoclonal antibodies to Dkk1 and sclerostin (AMG 785) have been developed as new treatments for osteoporosis (146, 147, 187). The anti-sclerostin antibodies, currently in phase 3 trials, appear to be most promising and may be a novel anabolic therapy for osteoporosis. If these antibodies do not have the drawback of the “anabolic window” of recombinant PTH (1-34 teriparatide), then they may replace the latter as the anabolic treatment agent of choice.
Conclusions
The current understanding of bone remodeling is primarily supported by the experimental results obtained from both in vivo and in vitro research. Many studies have been conducted on the generation and differentiation of different bone cells and involvement of these bone cells along with different stimuli on bone formation and bone remodeling. On the basis of these observations, many hypotheses have been proposed to understand the complex interactions of hormones, growth factors, and their receptors, and their autocrine, paracrine, and endocrine modes of signaling, which regulate physiological bone remodeling. This knowledge has led us from a period of treating metabolic bone diseases by classical endocrinology to one of using the growth factors or their inhibitors to treat these diseases. However, such a complex regulatory system, which involves numerous factors and their interactions, means that understanding of the behavior of the bone system is still fragmentary, and further knowledge is needed to maximize treatment options and to best target the growth factors.
Footnotes
No conflicts of interest, financial or otherwise, are declared by the author(s).
Author contributions: J.A.S. prepared figures; J.A.S. drafted manuscript; N.C.P. edited and revised manuscript; N.C.P. approved final version of manuscript.
References
- 1.Abu EO, Horner A, Kusec V, Triffitt JT, Compston JE. The localization of the functional glucocorticoid receptor alpha in human bone. J Clin Endocrinol Metab : 883–889, 2000. [DOI] [PubMed] [Google Scholar]
- 2.Alliston T, Choy L, Ducy P, Karsenty G, Derynck R. TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. EMBO J : 2254–2272, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Almeida M, Iyer S, Martin-Millan M, Bartell SM, Han L, Ambrogini E, Onal M, Xiong J, Weinstein RS, Jilka RL, O'Brien CA, Manolagas SC. Estrogen receptor-alpha signaling in osteoblast progenitors stimulates cortical bone accrual. J Clin Invest : 394–404,2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ammann P, Rizzoli R, Bonjour JP, Bourrin S, Meyer JM, Vassalli P, Garcia I. Transgenic mice expressing soluble tumor necrosis factor-receptor are protected against bone loss caused by estrogen deficiency. J Clin Invest : 1699–1703, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Anderson PH, Atkins GJ. The skeleton as an intracrine organ for vitamin D metabolism. Mol Aspects Med : 397–406, 2008. [DOI] [PubMed] [Google Scholar]
- 6.Anderson PH, Sawyer RK, Moore AJ, May BK, O'Loughlin PD, Morris HA. Vitamin D depletion induces RANKL-mediated osteoclastogenesis and bone loss in a rodent model. J Bone Miner Res : 1789–1797, 2008. [DOI] [PubMed] [Google Scholar]
- 7.Atkins GJ, Anderson PH, Findlay DM, Welldon KJ, Vincent C, Zannettino AC, O'Loughlin PD, Morris HA. Metabolism of vitamin D3 in human osteoblasts: evidence for autocrine and paracrine activities of 1 alpha,25-dihydroxyvitamin D3. Bone : 1517–1528, 2007. [DOI] [PubMed] [Google Scholar]
- 8.Bandyopadhyay A, Tsuji K, Cox K, Harfe BD, Rosen V, Tabin CJ. Genetic analysis of the roles of BMP2, BMP4, and BMP7 in limb patterning and skeletogenesis. PLoS Genet : e216, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bassett JH, Boyde A, Howell PG, Bassett RH, Galliford TM, Archanco M, Evans H, Lawson MA, Croucher P, St Germain DL, Galton VA, Williams GR. Optimal bone strength and mineralization requires the type 2 iodothyronine deiodinase in osteoblasts. Proc Natl Acad Sci USA : 7604–7609, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bassett JH, O'Shea PJ, Sriskantharajah S, Rabier B, Boyde A, Howell PG, Weiss RE, Roux JP, Malaval L, Clement-Lacroix P, Samarut J, Chassande O, Williams GR. Thyroid hormone excess rather than thyrotropin deficiency induces osteoporosis in hyperthyroidism. Mol Endocrinol : 1095–1107, 2007. [DOI] [PubMed] [Google Scholar]
- 11.Bedi B, Li JY, Tawfeek H, Baek KH, Adams J, Vangara SS, Chang MK, Kneissel M, Weitzmann MN, Pacifici R. Silencing of parathyroid hormone (PTH) receptor 1 in T cells blunts the bone anabolic activity of PTH. Proc Natl Acad Sci USA : 725–733, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bellido T, Saini V, Pajevic PD. Effects of PTH on osteocyte function. Bone : 250–257, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bennett CN, Longo KA, Wright WS, Suva LJ, Lane TF, Hankenson KD, MacDougald OA. Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc Natl Acad Sci USA : 3324–3329, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bennett CN, Ouyang H, Ma YL, Zeng Q, Gerin I, Sousa KM, Lane TF, Krishnan V, Hankenson KD, MacDougald OA. Wnt10b increases postnatal bone formation by enhancing osteoblast differentiation. J Bone Miner Res : 1924–1932, 2007. [DOI] [PubMed] [Google Scholar]
- 15.Benz DJ, Haussler MR, Thomas MA, Speelman B, Komm BS. High-affinity androgen binding and androgenic regulation of alpha 1(I)-procollagen and transforming growth factor-beta steady state messenger ribonucleic acid levels in human osteoblast-like osteosarcoma cells. Endocrinology : 2723–2730, 1991. [DOI] [PubMed] [Google Scholar]
- 16.Bikle D, Majumdar S, Laib A, Powell-Braxton L, Rosen C, Beamer W, Nauman E, Leary C, Halloran B. The skeletal structure of insulin-like growth factor I-deficient mice. J Bone Miner Res : 2320–2329, 2001. [DOI] [PubMed] [Google Scholar]
- 17.Bikle DD, Sakata T, Leary C, Elalieh H, Ginzinger D, Rosen CJ, Beamer W, Majumdar S, Halloran BP. Insulin-like growth factor I is required for the anabolic actions of parathyroid hormone on mouse bone. J Bone Miner Res : 1570–1578, 2002. [DOI] [PubMed] [Google Scholar]
- 18.Bochukova E, Schoenmakers N, Agostini M, Schoenmakers E, Rajanayagam O, Keogh JM, Henning E, Reinemund J, Gevers E, Sarri M, Downes K, Offiah A, Albanese A, Halsall D, Schwabe JW, Bain M, Lindley K, Muntoni F, Vargha-Khadem F, Dattani M, Farooqi IS, Gurnell M, Chatterjee K. A mutation in the thyroid hormone receptor alpha gene. N Engl J Med : 243–249, 2012. [DOI] [PubMed] [Google Scholar]
- 19.Britto JM, Fenton AJ, Holloway WR, Nicholson GC. Osteoblasts mediate thyroid hormone stimulation of osteoclastic bone resorption. Endocrinology : 169–176, 1994. [DOI] [PubMed] [Google Scholar]
- 20.Burkus JK, Transfeldt EE, Kitchel SH, Watkins RG, Balderston RA. Clinical and radiographic outcomes of anterior lumbar interbody fusion using recombinant human bone morphogenetic protein-2. Spine : 2396–2408, 2002. [DOI] [PubMed] [Google Scholar]
- 21.Canalis E. Clinical review 83: Mechanisms of glucocorticoid action in bone: implications to glucocorticoid-induced osteoporosis. J Clin Endocrinol Metab : 3441–3447, 1996. [DOI] [PubMed] [Google Scholar]
- 22.Carragee EJ, Hurwitz EL, Weiner BK. A critical review of recombinant human bone morphogenetic protein-2 trials in spinal surgery: emerging safety concerns and lessons learned. Spine J : 471–491, 2011. [DOI] [PubMed] [Google Scholar]
- 23.Casado-Diaz A, Santiago-Mora R, Quesada JM. The N- and C-terminal domains of parathyroid hormone-related protein affect differently the osteogenic and adipogenic potential of human mesenchymal stem cells. Exp Mol Med : 87–98, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cawthorn WP, Bree AJ, Yao Y, Du B, Hemati N, Martinez-Santibanez G, MacDougald OA. Wnt6, Wnt10a and Wnt10b inhibit adipogenesis and stimulate osteoblastogenesis through a beta-catenin-dependent mechanism. Bone : 477–489, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cenci S, Weitzmann MN, Roggia C, Namba N, Novack D, Woodring J, Pacifici R. Estrogen deficiency induces bone loss by enhancing T-cell production of TNF-alpha. J Clin Invest : 1229–1237, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chambers TJ, Athanasou NA, Fuller K. Effect of parathyroid hormone and calcitonin on the cytoplasmic spreading of isolated osteoclasts. J Endocrinol : 281–286, 1984. [DOI] [PubMed] [Google Scholar]
- 27.Chambers TJ, Chambers JC, Symonds J, Darby JA. The effect of human calcitonin on the cytoplasmic spreading of rat osteoclasts. J Clin Endocrinol Metab : 1080–1085, 1986. [DOI] [PubMed] [Google Scholar]
- 28.Chambers TJ, Magnus CJ. Calcitonin alters behaviour of isolated osteoclasts. J Pathol : 27–39, 1982. [DOI] [PubMed] [Google Scholar]
- 29.Chandra A, Lan S, Zhu J, Siclari VA, Qin L. Epidermal growth factor receptor (EGFR) signaling promotes proliferation and survival in osteoprogenitors by increasing early growth response 2 (EGR2) expression. J Biol Chem : 20488–20498, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen G, Deng C, Li YP. TGF-beta and BMP signaling in osteoblast differentiation and bone formation. Int J Biol Sci : 272–288, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chiang C, Chiu M, Moore AJ, Anderson PH, Ghasem-Zadeh A, McManus JF, Ma C, Seeman E, Clemens TL, Morris HA, Zajac JD, Davey RA. Mineralization and bone resorption are regulated by the androgen receptor in male mice. J Bone Miner Res : 621–631, 2009. [DOI] [PubMed] [Google Scholar]
- 32.Coe LM, Madathil SV, Casu C, Lanske B, Rivella S, Sitara D. FGF-23 is a negative regulator of prenatal and postnatal erythropoiesis. J Biol Chem : 9795–9810, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Courpron P, Meunier P, Vignon G. [Dynamics of bone remodeling explained by Harold Frost. Theory of the BMU (basic multicellular unit)]. Nouv Presse Med : 421–424, 1975. [PubMed] [Google Scholar]
- 34.Davideau JL, Papagerakis P, Hotton D, Lezot F, Berdal A. In situ investigation of vitamin D receptor, alkaline phosphatase, and osteocalcin gene expression in oro-facial mineralized tissues. Endocrinology : 3577–3585, 1996. [DOI] [PubMed] [Google Scholar]
- 35.Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell : 739–750, 2005. [DOI] [PubMed] [Google Scholar]
- 36.Dempster DW, Cosman F, Parisien M, Shen V, Lindsay R. Anabolic actions of parathyroid hormone on bone. Endocr Rev : 690–709, 1993. [DOI] [PubMed] [Google Scholar]
- 37.Dobnig H, Turner RT. The effects of programmed administration of human parathyroid hormone fragment (1–34) on bone histomorphometry and serum chemistry in rats. Endocrinology : 4607–4612, 1997. [DOI] [PubMed] [Google Scholar]
- 38.Dobnig H, Turner RT. Evidence that intermittent treatment with parathyroid hormone increases bone formation in adult rats by activation of bone lining cells. Endocrinology : 3632–3638, 1995. [DOI] [PubMed] [Google Scholar]
- 39.Erlebacher A, Derynck R. Increased expression of TGF-beta 2 in osteoblasts results in an osteoporosis-like phenotype. J Cell Biol : 195–210, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ernst M, Rodan GA. Increased activity of insulin-like growth factor (IGF) in osteoblastic cells in the presence of growth hormone (GH): positive correlation with the presence of the GH-induced IGF-binding protein BP-3. Endocrinology : 807–814, 1990. [DOI] [PubMed] [Google Scholar]
- 41.Falahati-Nini A, Riggs BL, Atkinson EJ, O'Fallon WM, Eastell R, Khosla S. Relative contributions of testosterone and estrogen in regulating bone resorption and formation in normal elderly men. J Clin Invest : 1553–1560, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Farley J, Dimai HP, Stilt-Coffing B, Farley P, Pham T, Mohan S. Calcitonin increases the concentration of insulin-like growth factors in serum-free cultures of human osteoblast-line cells. Calcif Tissue Int : 247–254, 2000. [DOI] [PubMed] [Google Scholar]
- 43.Finkelman RD, Mohan S, Linkhart TA, Abraham SM, Boussy JP, Baylink DJ. PTH stimulates the proliferation of TE-85 human osteosarcoma cells by a mechanism not involving either increased cAMP or increased secretion of IGF-I, IGF-II or TGF beta. Bone Miner : 89–100, 1992. [DOI] [PubMed] [Google Scholar]
- 44.Force T, Bonventre JV, Flannery MR, Gorn AH, Yamin M, Goldring SR. A cloned porcine renal calcitonin receptor couples to adenylyl cyclase and phospholipase C. Am J Physiol Renal Fluid Electrolyte Physiol : F1110–F1115, 1992. [DOI] [PubMed] [Google Scholar]
- 45.Freitas FR, Moriscot AS, Jorgetti V, Soares AG, Passarelli M, Scanlan TS, Brent GA, Bianco AC, Gouveia CH. Spared bone mass in rats treated with thyroid hormone receptor TR beta-selective compound GC-1. Am J Physiol Endocrinol Metab : E1135–E1141, 2003. [DOI] [PubMed] [Google Scholar]
- 46.Frenkel B, Hong A, Baniwal SK, Coetzee GA, Ohlsson C, Khalid O, Gabet Y. Regulation of adult bone turnover by sex steroids. J Cell Physiol : 305–310, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gardella TJ, Juppner H. Interaction of PTH and PTHrP with their receptors. Rev Endocr Metab Disord : 317–329, 2000. [DOI] [PubMed] [Google Scholar]
- 48.Geiser AG, Zeng QQ, Sato M, Helvering LM, Hirano T, Turner CH. Decreased bone mass and bone elasticity in mice lacking the transforming growth factor-beta1 gene. Bone : 87–93, 1998. [DOI] [PubMed] [Google Scholar]
- 49.Gennari C. Differential effect of glucocorticoids on calcium absorption and bone mass. Br J Rheumatol , Suppl 2: 11–14, 1993. [DOI] [PubMed] [Google Scholar]
- 50.Glass DA 2nd Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, McMahon AP, Lang RA, Karsenty G. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell : 751–764, 2005. [DOI] [PubMed] [Google Scholar]
- 51.Gogakos AI, Duncan Bassett JH, Williams GR. Thyroid and bone. Arch Biochem Biophys : 129–136, 2010. [DOI] [PubMed] [Google Scholar]
- 52.Goltzman D. Studies on the mechanisms of the skeletal anabolic action of endogenous and exogenous parathyroid hormone. Arch Biochem Biophys : 218–224, 2008. [DOI] [PubMed] [Google Scholar]
- 53.Gooi JH, Chia LY, Walsh NC, Karsdal MA, Quinn JM, Martin TJ, Sims NA. Decline in calcitonin receptor expression in osteocytes with age. J Endocrinol : 181–191, 2014. [DOI] [PubMed] [Google Scholar]
- 54.Gooi JH, Pompolo S, Karsdal MA, Kulkarni NH, Kalajzic I, McAhren SH, Han B, Onyia JE, Ho PW, Gillespie MT, Walsh NC, Chia LY, Quinn JM, Martin TJ, Sims NA. Calcitonin impairs the anabolic effect of PTH in young rats and stimulates expression of sclerostin by osteocytes. Bone : 1486–1497, 2010. [DOI] [PubMed] [Google Scholar]
- 55.Govender S, Csimma C, Genant HK, Valentin-Opran A, Amit Y, Arbel R, Aro H, Atar D, Bishay M, Borner MG, Chiron P, Choong P, Cinats J, Courtenay B, Feibel R, Geulette B, Gravel C, Haas N, Raschke M, Hammacher E, van der Velde D, Hardy P, Holt M, Josten C, Ketterl RL, Lindeque B, Lob G, Mathevon H, McCoy G, Marsh D, Miller R, Munting E, Oevre S, Nordsletten L, Patel A, Pohl A, Rennie W, Reynders P, Rommens PM, Rondia J, Rossouw WC, Daneel PJ, Ruff S, Ruter A, Santavirta S, Schildhauer TA, Gekle C, Schnettler R, Segal D, Seiler H, Snowdowne RB, Stapert J, Taglang G, Verdonk R, Vogels L, Weckbach A, Wentzensen A, Wisniewski T. Recombinant human bone morphogenetic protein-2 for treatment of open tibial fractures: a prospective, controlled, randomized study of four hundred and fifty patients. J Bone Joint Surg Am : 2123–2134, 2002. [DOI] [PubMed] [Google Scholar]
- 56.Guevarra MS, Yeh JK, Castro Magana M, Aloia JF. Synergistic effect of parathyroid hormone and growth hormone on trabecular and cortical bone formation in hypophysectomized rats. Hormone Res Paediatr : 248–257, 2010. [DOI] [PubMed] [Google Scholar]
- 57.Guo J, Liu M, Yang D, Bouxsein ML, Saito H, Galvin RJ, Kuhstoss SA, Thomas CC, Schipani E, Baron R, Bringhurst FR, Kronenberg HM. Suppression of Wnt signaling by Dkk1 attenuates PTH-mediated stromal cell response and new bone formation. Cell Metab : 161–171, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hamidouche Z, Fromigue O, Nuber U, Vaudin P, Pages JC, Ebert R, Jakob F, Miraoui H, Marie PJ. Autocrine fibroblast growth factor 18 mediates dexamethasone-induced osteogenic differentiation of murine mesenchymal stem cells. J Cell Physiol : 509–515, 2010. [DOI] [PubMed] [Google Scholar]
- 59.Hayden JM, Mohan S, Baylink DJ. The insulin-like growth factor system and the coupling of formation to resorption. Bone : 93S–98S, 1995. [DOI] [PubMed] [Google Scholar]
- 60.Hildreth BE 3rd Werbeck JL, Thudi NK, Deng X, Rosol TJ, Toribio RE. PTHrP 1-141 and 1-86 increase in vitro bone formation. J Surg Res : e9–e17, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, Spelsberg TC, Khosla S. Stimulation of osteoprotegerin ligand and inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage cells: potential paracrine mechanisms of glucocorticoid-induced osteoporosis. Endocrinology : 4382–4389, 1999. [DOI] [PubMed] [Google Scholar]
- 62.Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, Deng L, Clemens TL, Williams BO. Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem : 21162–21168, 2005. [DOI] [PubMed] [Google Scholar]
- 63.Houde N, Chamoux E, Bisson M, Roux S. Transforming growth factor-beta1 (TGF-beta1) induces human osteoclast apoptosis by up-regulating Bim. J Biol Chem : 23397–23404, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Huang Z, Ren PG, Ma T, Smith RL, Goodman SB. Modulating osteogenesis of mesenchymal stem cells by modifying growth factor availability. Cytokine : 305–310, 2010. [DOI] [PubMed] [Google Scholar]
- 65.Ikegame M, Rakopoulos M, Zhou H, Houssami S, Martin TJ, Moseley JM, Findlay DM. Calcitonin receptor isoforms in mouse and rat osteoclasts. J Bone Miner Res : 59–65, 1995. [DOI] [PubMed] [Google Scholar]
- 66.Imanishi Y, Kobayashi K, Kawata T, Inaba M, Nishizawa Y. Regulatory mechanisms of circulating fibroblast growth factor 23 in parathyroid diseases. Ther Apher Dial , Suppl 1: S32–S37, 2007. [DOI] [PubMed] [Google Scholar]
- 67.Inzerillo AM, Zaidi M, Huang CL. Calcitonin: the other thyroid hormone. Thyroid : 791–798, 2002. [DOI] [PubMed] [Google Scholar]
- 68.Ishida Y, Heersche JN. Glucocorticoid-induced osteoporosis: both in vivo and in vitro concentrations of glucocorticoids higher than physiological levels attenuate osteoblast differentiation. J Bone Miner Res : 1822–1826, 1998. [DOI] [PubMed] [Google Scholar]
- 69.Jacob AL, Smith C, Partanen J, Ornitz DM. Fibroblast growth factor receptor 1 signaling in the osteo-chondrogenic cell lineage regulates sequential steps of osteoblast maturation. Dev Biol : 315–328, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jilka RL, O'Brien CA, Ali AA, Roberson PK, Weinstein RS, Manolagas SC. Intermittent PTH stimulates periosteal bone formation by actions on post-mitotic preosteoblasts. Bone : 275–286, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Jilka RL, Weinstein RS, Bellido T, Roberson P, Parfitt AM, Manolagas SC. Increased bone formation by prevention of osteoblast apoptosis with parathyroid hormone. J Clin Invest : 439–446, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Juppner H, Abou-Samra AB, Freeman M, Kong XF, Schipani E, Richards J, Kolakowski LF Jr, Hock J, Potts JT Jr, Kronenberg HM, et al. A G protein-linked receptor for parathyroid hormone and parathyroid hormone-related peptide. Science : 1024–1026, 1991. [DOI] [PubMed] [Google Scholar]
- 73.Kanatani M, Sugimoto T, Sowa H, Kobayashi T, Kanzawa M, Chihara K. Thyroid hormone stimulates osteoclast differentiation by a mechanism independent of RANKL-RANK interaction. J Cell Physiol : 17–25, 2004. [DOI] [PubMed] [Google Scholar]
- 74.Karaplis AC, Luz A, Glowacki J, Bronson RT, Tybulewicz VL, Kronenberg HM, Mulligan RC. Lethal skeletal dysplasia from targeted disruption of the parathyroid hormone-related peptide gene. Genes Dev : 277–289, 1994. [DOI] [PubMed] [Google Scholar]
- 75.Kartsogiannis V, Moseley J, McKelvie B, Chou ST, Hards DK, Ng KW, Martin TJ, Zhou H. Temporal expression of PTHrP during endochondral bone formation in mouse and intramembranous bone formation in an in vivo rabbit model. Bone : 385–392, 1997. [DOI] [PubMed] [Google Scholar]
- 76.Kasperk CH, Faehling K, Borcsok I, Ziegler R. Effects of androgens on subpopulations of the human osteosarcoma cell line SaOS2. Calcif Tissue Int : 376–382, 1996. [DOI] [PubMed] [Google Scholar]
- 77.Kato H, Ochiai-Shino H, Onodera S, Saito A, Shibahara T, Azuma T. Promoting effect of 1,25(OH)2 vitamin D3 in osteogenic differentiation from induced pluripotent stem cells to osteocyte-like cells. Open Biol : 140201, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass DA 2nd Hartmann C, Li L, Hwang TH, Brayton CF, Lang RA, Karsenty G, Chan L. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol : 303–314, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Katznelson L, Finkelstein JS, Schoenfeld DA, Rosenthal DI, Anderson EJ, Klibanski A. Increase in bone density and lean body mass during testosterone administration in men with acquired hypogonadism. J Clin Endocrinol Metab : 4358–4365, 1996. [DOI] [PubMed] [Google Scholar]
- 80.Kawano H, Sato T, Yamada T, Matsumoto T, Sekine K, Watanabe T, Nakamura T, Fukuda T, Yoshimura K, Yoshizawa T, Aihara K, Yamamoto Y, Nakamichi Y, Metzger D, Chambon P, Nakamura K, Kawaguchi H, Kato S. Suppressive function of androgen receptor in bone resorption. Proc Natl Acad Sci USA : 9416–9421, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kawata T, Imanishi Y, Kobayashi K, Miki T, Arnold A, Inaba M, Nishizawa Y. Parathyroid hormone regulates fibroblast growth factor-23 in a mouse model of primary hyperparathyroidism. J Am Soc Nephrol : 2683–2688, 2007. [DOI] [PubMed] [Google Scholar]
- 82.Keller J, Catala-Lehnen P, Huebner AK, Jeschke A, Heckt T, Lueth A, Krause M, Koehne T, Albers J, Schulze J, Schilling S, Haberland M, Denninger H, Neven M, Hermans-Borgmeyer I, Streichert T, Breer S, Barvencik F, Levkau B, Rathkolb B, Wolf E, Calzada-Wack J, Neff F, Gailus-Durner V, Fuchs H, de Angelis MH, Klutmann S, Tsourdi E, Hofbauer LC, Kleuser B, Chun J, Schinke T, Amling M. Calcitonin controls bone formation by inhibiting the release of sphingosine 1-phosphate from osteoclasts. Nat Commun : 5215, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kennell JA, MacDougald OA. Wnt signaling inhibits adipogenesis through beta-catenin-dependent and -independent mechanisms. J Biol Chem : 24004–24010, 2005. [DOI] [PubMed] [Google Scholar]
- 84.Khosla S. Minireview: the OPG/RANKL/RANK system. Endocrinology : 5050–5055, 2001. [DOI] [PubMed] [Google Scholar]
- 85.Kimble RB, Srivastava S, Ross FP, Matayoshi A, Pacifici R. Estrogen deficiency increases the ability of stromal cells to support murine osteoclastogenesis via an interleukin-1and tumor necrosis factor-mediated stimulation of macrophage colony-stimulating factor production. J Biol Chem : 28890–28897, 1996. [DOI] [PubMed] [Google Scholar]
- 86.Kogawa M, Anderson PH, Findlay DM, Morris HA, Atkins GJ. The metabolism of 25-(OH)vitamin D3 by osteoclasts and their precursors regulates the differentiation of osteoclasts. J Steroid Biochem Mol Biol : 277–280, 2010. [DOI] [PubMed] [Google Scholar]
- 87.Kogawa M, Findlay DM, Anderson PH, Ormsby R, Vincent C, Morris HA, Atkins GJ. Osteoclastic metabolism of 25(OH)-vitamin D3: a potential mechanism for optimization of bone resorption. Endocrinology : 4613–4625, 2010. [DOI] [PubMed] [Google Scholar]
- 88.Kolek OI, Hines ER, Jones MD, LeSueur LK, Lipko MA, Kiela PR, Collins JF, Haussler MR, Ghishan FK. 1Alpha,25-Dihydroxyvitamin D3 upregulates FGF23 gene expression in bone: the final link in a renal-gastrointestinal-skeletal axis that controls phosphate transport. Am J Physiol Gastrointest Liver Physiol : G1036–G1042, 2005. [DOI] [PubMed] [Google Scholar]
- 89.Kondo T, Kitazawa R, Maeda S, Kitazawa S. 1 Alpha,25 dihydroxyvitamin D3 rapidly regulates the mouse osteoprotegerin gene through dual pathways. J Bone Miner Res : 1411–1419, 2004. [DOI] [PubMed] [Google Scholar]
- 90.Kostenuik PJ, Harris J, Halloran BP, Turner RT, Morey-Holton ER, Bikle DD. Skeletal unloading causes resistance of osteoprogenitor cells to parathyroid hormone and to insulin-like growth factor-I. J Bone Miner Res : 21–31, 1999. [DOI] [PubMed] [Google Scholar]
- 91.Kousteni S, Bellido T, Plotkin LI, O'Brien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC. Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell : 719–730, 2001. [PubMed] [Google Scholar]
- 92.Kousteni S, Han L, Chen JR, Almeida M, Plotkin LI, Bellido T, Manolagas SC. Kinase-mediated regulation of common transcription factors accounts for the bone-protective effects of sex steroids. J Clin Invest : 1651–1664, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kramer I, Halleux C, Keller H, Pegurri M, Gooi JH, Weber PB, Feng JQ, Bonewald LF, Kneissel M. Osteocyte Wnt/beta-catenin signaling is required for normal bone homeostasis. Mol Cell Biol : 3071–3085, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Krause C, Korchynskyi O, de Rooij K, Weidauer SE, de Gorter DJ, van Bezooijen RL, Hatsell S, Economides AN, Mueller TD, Lowik CW, ten Dijke P. Distinct modes of inhibition by sclerostin on bone morphogenetic protein and Wnt signaling pathways. J Biol Chem : 41614–41626, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt signaling. J Clin Invest : 1202–1209, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kronenberg HM. PTHrP and skeletal development. Ann NY Acad Sci : 1–13, 2006. [DOI] [PubMed] [Google Scholar]
- 97.Kudo TA, Kanetaka H, Watanabe A, Okumoto A, Asano M, Zhang Y, Zhao F, Kano M, Shimizu Y, Tamura S, Hayashi H. Investigating bone morphogenetic protein (BMP) signaling in a newly established human cell line expressing BMP receptor type II Tohoku. J Exp Med : 121–129, 2010. [DOI] [PubMed] [Google Scholar]
- 98.Kuzma M, Kuzmova Z, Zelinkova Z, Killinger Z, Vanuga P, Lazurova I, Tomkova S, Payer J. Impact of the growth hormone replacement on bone status in growth hormone deficient adults. Growth Hormone : 22–28, 2014. [DOI] [PubMed] [Google Scholar]
- 99.Lanske B, Divieti P, Kovacs CS, Pirro A, Landis WJ, Krane SM, Bringhurst FR, Kronenberg HM. The parathyroid hormone (PTH)/PTH-related peptide receptor mediates actions of both ligands in murine bone. Endocrinology : 5194–5204, 1998. [DOI] [PubMed] [Google Scholar]
- 100.Lanske B, Karaplis AC, Lee K, Luz A, Vortkamp A, Pirro A, Karperien M, Defize LH, Ho C, Mulligan RC, Abou-Samra AB, Juppner H, Segre GV, Kronenberg HM. PTH/PTHrP receptor in early development and Indian hedgehog-regulated bone growth. Science : 663–666, 1996. [DOI] [PubMed] [Google Scholar]
- 101.Lazowski DA, Fraher LJ, Hodsman A, Steer B, Modrowski D, Han VK. Regional variation of insulin-like growth factor-I gene expression in mature rat bone and cartilage. Bone : 563–576, 1994. [DOI] [PubMed] [Google Scholar]
- 102.Leaffer D, Sweeney M, Kellerman LA, Avnur Z, Krstenansky JL, Vickery BH, Caulfield JP. Modulation of osteogenic cell ultrastructure by RS-23581, an analog of human parathyroid hormone (PTH)-related peptide-(1–34), and bovine PTH-(1–34). Endocrinology : 3624–3631, 1995. [DOI] [PubMed] [Google Scholar]
- 103.Leder BZ, O'Dea LS, Zanchetta JR, Kumar P, Banks K, McKay K, Lyttle CR, Hattersley G. Effects of abaloparatide, a human parathyroid hormone-related peptide analog, on bone mineral density in postmenopausal women with osteoporosis. J Clin Endocrinol Metab : 697–706, 2015. [DOI] [PubMed] [Google Scholar]
- 104.Lee H, Finkelstein JS, Miller M, Comeaux SJ, Cohen RI, Leder BZ. Effects of selective testosterone and estradiol withdrawal on skeletal sensitivity to parathyroid hormone in men. J Clin Endocrinol Metab : 1069–1075, 2006. [DOI] [PubMed] [Google Scholar]
- 105.Lee SK, Lorenzo JA. Parathyroid hormone stimulates TRANCE and inhibits osteoprotegerin messenger ribonucleic acid expression in murine bone marrow cultures: correlation with osteoclast-like cell formation. Endocrinology : 3552–3561, 1999. [DOI] [PubMed] [Google Scholar]
- 106.Li X, Qin L, Bergenstock M, Bevelock LM, Novack DV, Partridge NC. Parathyroid hormone stimulates osteoblastic expression of MCP-1 to recruit and increase the fusion of pre/osteoclasts. J Biol Chem : 33098–33106, 2007. [DOI] [PubMed] [Google Scholar]
- 107.Li Y, He X, Olauson H, Larsson TE, Lindgren U. FGF23 affects the lineage fate determination of mesenchymal stem cells. Calc Tissue Int : 556–564, 2013. [DOI] [PubMed] [Google Scholar]
- 108.Li YC, Pirro AE, Amling M, Delling G, Baron R, Bronson R, Demay MB. Targeted ablation of the vitamin D receptor: an animal model of vitamin D-dependent rickets type II with alopecia. Proc Natl Acad Sci USA : 9831–9835, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lin HY, Harris TL, Flannery MS, Aruffo A, Kaji EH, Gorn A, Kolakowski LF Jr,. Lodish HF, and Goldring SR. Expression cloning of an adenylate cyclase-coupled calcitonin receptor. Science : 1022–1024, 1991. [DOI] [PubMed] [Google Scholar]
- 110.Liu Z, Kennedy OD, Cardoso L, Basta-Pljakic J, Partridge NC, Schaffler MB, Rosen CJ, Yakar S. DMP-1-mediated Ghr gene recombination compromises skeletal development and impairs skeletal response to intermittent PTH. FASEB J : 635–652, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Locklin RM, Khosla S, Turner RT, Riggs BL. Mediators of the biphasic responses of bone to intermittent and continuously administered parathyroid hormone. J Cell Biochem : 180–190, 2003. [DOI] [PubMed] [Google Scholar]
- 112.Lomri A, de Pollak C, Sebag M, Goltzman D, Kremer R, Marie PJ. Expression of parathyroid hormone-related peptide (PTHrP) and PTH/PTHrP receptor in newborn human calvaria osteoblastic cells. Eur J Endocrinol : 640–648, 1997. [DOI] [PubMed] [Google Scholar]
- 113.Lukert BP. Osteoporosis: prevention and treatment. Compr Ther : 36–42, 1990. [PubMed] [Google Scholar]
- 114.Lukert BP, Raisz LG. Glucocorticoid-induced osteoporosis: pathogenesis and management. Ann Intern Med : 352–364, 1990. [DOI] [PubMed] [Google Scholar]
- 115.Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizoguchi T, Kikuchi Y, Takada I, Kato S, Kani S, Nishita M, Marumo K, Martin TJ, Minami Y, Takahashi N. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med : 405–412, 2012. [DOI] [PubMed] [Google Scholar]
- 116.Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev : 115–137, 2000. [DOI] [PubMed] [Google Scholar]
- 117.Manolagas SC, Jilka RL. Bone marrow, cytokines, and bone remodeling. Emerging insights into the pathophysiology of osteoporosis. N Engl J Med : 305–311, 1995. [DOI] [PubMed] [Google Scholar]
- 118.Martin-Millan M, Almeida M, Ambrogini E, Han L, Zhao H, Weinstein RS, Jilka RL, O'Brien CA, Manolagas SC. The estrogen receptor-alpha in osteoclasts mediates the protective effects of estrogens on cancellous but not cortical bone. Mol Endocrinol : 323–334, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Martin A, David V, Quarles LD. Regulation and function of the FGF23/klotho endocrine pathways. Physiol Rev : 131–155, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Martin I, Muraglia A, Campanile G, Cancedda R, Quarto R. Fibroblast growth factor-2 supports ex vivo expansion and maintenance of osteogenic precursors from human bone marrow. Endocrinology : 4456–4462, 1997. [DOI] [PubMed] [Google Scholar]
- 121.Martin T, Gooi JH, Sims NA. Molecular mechanisms in coupling of bone formation to resorption. Crit Rev Eukaryot Gene Expr : 73–88, 2009. [DOI] [PubMed] [Google Scholar]
- 122.Maruyama T, Mirando AJ, Deng CX, Hsu W. The balance of WNT and FGF signaling influences mesenchymal stem cell fate during skeletal development. Sci Signal : ra40, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Melville KM, Kelly NH, Khan SA, Schimenti JC, Ross FP, Main RP, van der Meulen MC. Female mice lacking estrogen receptor-alpha in osteoblasts have compromised bone mass and strength. J Bone Miner Res : 370–379, 2014. [DOI] [PubMed] [Google Scholar]
- 124.Miao D, He B, Jiang Y, Kobayashi T, Soroceanu MA, Zhao J, Su H, Tong X, Amizuka N, Gupta A, Genant HK, Kronenberg HM, Goltzman D, Karaplis AC. Osteoblast-derived PTHrP is a potent endogenous bone anabolic agent that modifies the therapeutic efficacy of administered PTH 1–34. J Clin Invest : 2402–2411, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Mishina Y, Starbuck MW, Gentile MA, Fukuda T, Kasparcova V, Seedor JG, Hanks MC, Amling M, Pinero GJ, Harada S, Behringer RR. Bone morphogenetic protein type IA receptor signaling regulates postnatal osteoblast function and bone remodeling. J Biol Chem : 27560–27566, 2004. [DOI] [PubMed] [Google Scholar]
- 126.Mohan S, Baylink DJ. Insulin-like growth factor system components and the coupling of bone formation to resorption. Horm Res , Suppl 1: 59–62, 1996. [DOI] [PubMed] [Google Scholar]
- 127.Mohan S, Kutilek S, Zhang C, Shen HG, Kodama Y, Srivastava AK, Wergedal JE, Beamer WG, Baylink DJ. Comparison of bone formation responses to parathyroid hormone(1-34), (1-31), and (2-34) in mice. Bone : 471–478, 2000. [DOI] [PubMed] [Google Scholar]
- 128.Monfoulet LE, Rabier B, Dacquin R, Anginot A, Photsavang J, Jurdic P, Vico L, Malaval L, Chassande O. Thyroid hormone receptor beta mediates thyroid hormone effects on bone remodeling and bone mass. J Bone Miner Res : 2036–2044, 2011. [DOI] [PubMed] [Google Scholar]
- 129.Monroe DG, McGee-Lawrence ME, Oursler MJ, Westendorf JJ. Update on Wnt signaling in bone cell biology and bone disease. Gene : 1–18, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Montero A, Okada Y, Tomita M, Ito M, Tsurukami H, Nakamura T, Doetschman T, Coffin JD, Hurley MM. Disruption of the fibroblast growth factor-2 gene results in decreased bone mass and bone formation. J Clin Invest : 1085–1093, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Moran C, Schoenmakers N, Agostini M, Schoenmakers E, Offiah A, Kydd A, Kahaly G, Mohr-Kahaly S, Rajanayagam O, Lyons G, Wareham N, Halsall D, Dattani M, Hughes S, Gurnell M, Park SM, Chatterjee K. An adult female with resistance to thyroid hormone mediated by defective thyroid hormone receptor alpha. J Clin Endocrinol Metab : 4254–4261, 2013. [DOI] [PubMed] [Google Scholar]
- 132.Morel G, Chavassieux P, Barenton B, Dubois PM, Meunier PJ, Boivin G. Evidence for a direct effect of growth hormone on osteoblasts. Cell Tissue Res : 279–286, 1993. [DOI] [PubMed] [Google Scholar]
- 133.Morris HA, Turner AG, Anderson PH. Vitamin-D regulation of bone mineralization and remodelling during growth. Front Biosci : 677–689, 2012. [DOI] [PubMed] [Google Scholar]
- 134.Mundy GR, Shapiro JL, Bandelin JG, Canalis EM, Raisz LG. Direct stimulation of bone resorption by thyroid hormones. J Clin Invest : 529–534, 1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Murphy E, Williams GR. The thyroid and the skeleton. Clin Endocrinol (Oxf) : 285–298, 2004. [DOI] [PubMed] [Google Scholar]
- 136.Naganawa T, Xiao L, Coffin JD, Doetschman T, Sabbieti MG, Agas D, Hurley MM. Reduced expression and function of bone morphogenetic protein-2 in bones of Fgf2 null mice. J Cell Biochem : 1975–1988, 2008. [DOI] [PubMed] [Google Scholar]
- 137.Nishida S, Yamaguchi A, Tanizawa T, Endo N, Mashiba T, Uchiyama Y, Suda T, Yoshiki S, Takahashi HE. Increased bone formation by intermittent parathyroid hormone administration is due to the stimulation of proliferation and differentiation of osteoprogenitor cells in bone marrow. Bone : 717–723, 1994. [DOI] [PubMed] [Google Scholar]
- 138.Nishiyama K, Sugimoto T, Kaji H, Kanatani M, Kobayashi T, Chihara K. Stimulatory effect of growth hormone on bone resorption and osteoclast differentiation. Endocrinology : 35–41, 1996. [DOI] [PubMed] [Google Scholar]
- 139.Noel D, Gazit D, Bouquet C, Apparailly F, Bony C, Plence P, Millet V, Turgeman G, Perricaudet M, Sany J, Jorgensen C. Short-term BMP-2 expression is sufficient for in vivo osteochondral differentiation of mesenchymal stem cells. Stem Cells : 74–85, 2004. [DOI] [PubMed] [Google Scholar]
- 140.Notini AJ, McManus JF, Moore A, Bouxsein M, Jimenez M, Chiu WS, Glatt V, Kream BE, Handelsman DJ, Morris HA, Zajac JD, Davey RA. Osteoblast deletion of exon 3 of the androgen receptor gene results in trabecular bone loss in adult male mice. J Bone Miner Res : 347–356, 2007. [DOI] [PubMed] [Google Scholar]
- 141.O'Brien CA, Plotkin LI, Galli C, Goellner JJ, Gortazar AR, Allen MR, Robling AG, Bouxsein M, Schipani E, Turner CH, Jilka RL, Weinstein RS, Manolagas SC, Bellido T. Control of bone mass and remodeling by PTH receptor signaling in osteocytes. PLoS One : e2942, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Ohbayashi N, Shibayama M, Kurotaki Y, Imanishi M, Fujimori T, Itoh N, Takada S. FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis. Genes Dev : 870–879, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ohlsson C, Vidal O. Effects of growth hormone and insulin-like growth factors on human osteoblasts. Eur J Clin Invest : 184–186, 1998. [DOI] [PubMed] [Google Scholar]
- 144.Okubo Y, Bessho K, Fujimura K, Kusumoto K, Ogawa Y, Iizuka T. Effect of elcatonin on osteoinduction by recombinant human bone morphogenetic protein-2. Biochem Biophys Res Commun : 317–321, 2000. [DOI] [PubMed] [Google Scholar]
- 145.Pacifici R, Brown C, Puscheck E, Friedrich E, Slatopolsky E, Maggio D, McCracken R, Avioli LV. Effect of surgical menopause and estrogen replacement on cytokine release from human blood mononuclear cells. Proc Natl Acad Sci USA : 5134–5138, 1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Padhi D, Allison M, Kivitz AJ, Gutierrez MJ, Stouch B, Wang C, Jang G. Multiple doses of sclerostin antibody romosozumab in healthy men and postmenopausal women with low bone mass: a randomized, double-blind, placebo-controlled study. J Clin Pharmacol : 168–178, 2014. [DOI] [PubMed] [Google Scholar]
- 147.Padhi D, Jang G, Stouch B, Fang L, Posvar E. Single-dose, placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. J Bone Miner Res : 19–26, 2011. [DOI] [PubMed] [Google Scholar]
- 148.Paic F, Igwe JC, Nori R, Kronenberg MS, Franceschetti T, Harrington P, Kuo L, Shin DG, Rowe DW, Harris SE, Kalajzic I. Identification of differentially expressed genes between osteoblasts and osteocytes. Bone : 682–692, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Panda DK, Miao D, Tremblay ML, Sirois J, Farookhi R, Hendy GN, Goltzman D. Targeted ablation of the 25-hydroxyvitamin D 1alpha -hydroxylase enzyme: evidence for skeletal, reproductive, and immune dysfunction. Proc Natl Acad Sci USA : 7498–7503, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Parfitt AM. The coupling of bone formation to bone resorption: a critical analysis of the concept and of its relevance to the pathogenesis of osteoporosis. Metab Bone Dis Relat Res : 1–6, 1982. [DOI] [PubMed] [Google Scholar]
- 151.Partridge NC, Alcorn D, Michelangeli VP, Kemp BE, Ryan GB, Martin TJ. Functional properties of hormonally responsive cultured normal and malignant rat osteoblastic cells. Endocrinology : 213–219, 1981. [DOI] [PubMed] [Google Scholar]
- 152.Pearce G, Tabensky DA, Delmas PD, Baker HW, Seeman E. Corticosteroid-induced bone loss in men. J Clin Endocrinol Metab : 801–806, 1998. [DOI] [PubMed] [Google Scholar]
- 153.Pereira RM, Carvalho JF, Canalis E. Glucocorticoid-induced osteoporosis in rheumatic diseases. Clinics (Sao Paulo) : 1197–1205, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Pereira RM, Delany AM, Canalis E. Cortisol inhibits the differentiation and apoptosis of osteoblasts in culture. Bone : 484–490, 2001. [DOI] [PubMed] [Google Scholar]
- 155.Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, Manolagas SC, Bellido T. Prevention of osteocyte and osteoblast apoptosis by bisphosphonates and calcitonin. J Clin Invest : 1363–1374, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Qiu T, Wu X, Zhang F, Clemens TL, Wan M, Cao X. TGF-beta type II receptor phosphorylates PTH receptor to integrate bone remodelling signalling. Nat Cell Biol : 224–234, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, Kirilov M, Mandic V, Takacz A, Schmidt-Ullrich R, Ostermay S, Schinke T, Spanbroek R, Zaiss MM, Angel PE, Lerner UH, David JP, Reichardt HM, Amling M, Schutz G, Tuckermann JP. Glucocorticoids suppress bone formation by attenuating osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab : 517–531, 2010. [DOI] [PubMed] [Google Scholar]
- 158.Rhee Y, Lee EY, Lezcano V, Ronda AC, Condon KW, Allen MR, Plotkin LI, Bellido T. Resorption controls bone anabolism driven by parathyroid hormone (PTH) receptor signaling in osteocytes. J Biol Chem : 29809–29820, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Rosenfeld RG, Rosenbloom AL, Guevara-Aguirre J. Growth hormone (GH) insensitivity due to primary GH receptor deficiency. Endocr Rev : 369–390, 1994. [DOI] [PubMed] [Google Scholar]
- 160.Russo D, Battaglia Y. Clinical significance of FGF-23 in patients with CKD. Int J Nephrol : 364890, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Sabbieti MG, Agas D, Xiao L, Marchetti L, Coffin JD, Doetschman T, Hurley MM. Endogenous FGF-2 is critically important in PTH anabolic effects on bone. J Cell Physiol : 143–151, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Sapir-Koren R, Livshits G. Bone mineralization is regulated by signaling cross talk between molecular factors of local and systemic origin: the role of fibroblast growth factor 23. Biofactors : 555–556, 2014. [DOI] [PubMed] [Google Scholar]
- 163.Scutt A, Duvos C, Lauber J, Mayer H. Time-dependent effects of parathyroid hormone and prostaglandin E2 on DNA synthesis by periosteal cells from embryonic chick calvaria. Calcif Tissue Int : 208–215, 1994. [DOI] [PubMed] [Google Scholar]
- 164.Shen B, Wei A, Whittaker S, Williams LA, Tao H, Ma DD, Diwan AD. The role of BMP-7 in chondrogenic and osteogenic differentiation of human bone marrow multipotent mesenchymal stromal cells in vitro. J Cell Biochem : 406–416, 2010. [DOI] [PubMed] [Google Scholar]
- 165.Shevde NK, Bendixen AC, Dienger KM, Pike JW. Estrogens suppress RANK ligand-induced osteoclast differentiation via a stromal cell independent mechanism involving c-Jun repression. Proc Natl Acad Sci USA : 7829–7834, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Shimada T, Muto T, Urakawa I, Yoneya T, Yamazaki Y, Okawa K, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T. Mutant FGF-23 responsible for autosomal dominant hypophosphatemic rickets is resistant to proteolytic cleavage and causes hypophosphatemia in vivo. Endocrinology : 3179–3182, 2002. [DOI] [PubMed] [Google Scholar]
- 167.Shimada T, Urakawa I, Yamazaki Y, Hasegawa H, Hino R, Yoneya T, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T. FGF-23 transgenic mice demonstrate hypophosphatemic rickets with reduced expression of sodium phosphate cotransporter type IIa. Biochem Biophys Res Commun : 409–414, 2004. [DOI] [PubMed] [Google Scholar]
- 168.Sims NA, Clement-Lacroix P, Da Ponte F, Bouali Y, Binart N, Moriggl R, Goffin V, Coschigano K, Gaillard-Kelly M, Kopchick J, Baron R, Kelly PA. Bone homeostasis in growth hormone receptor-null mice is restored by IGF-I but independent of Stat5. J Clin Invest : 1095–1103, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Sinnesael M, Claessens F, Laurent M, Dubois V, Boonen S, Deboel L, Vanderschueren D. Androgen receptor (AR) in osteocytes is important for the maintenance of male skeletal integrity: evidence from targeted AR disruption in mouse osteocytes. J Bone Miner Res : 2535–2543, 2012. [DOI] [PubMed] [Google Scholar]
- 170.Sitara D, Kim S, Razzaque MS, Bergwitz C, Taguchi T, Schuler C, Erben RG, Lanske B. Genetic evidence of serum phosphate-independent functions of FGF-23 on bone. PLoS Genet : e1000154, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Sjogren K, Bohlooly YM, Olsson B, Coschigano K, Tornell J, Mohan S, Isaksson OG, Baumann G, Kopchick J, Ohlsson C. Disproportional skeletal growth and markedly decreased bone mineral content in growth hormone receptor −/− mice. Biochem Biophys Res Commun : 603–608, 2000. [DOI] [PubMed] [Google Scholar]
- 172.Somjen D, Binderman I, Schluter KD, Wingender E, Mayer H, Kaye AM. Stimulation by defined parathyroid hormone fragments of cell proliferation in skeletal-derived cell cultures. Biochem J : 781–785, 1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Stevens JR, Miranda-Carboni GA, Singer MA, Brugger SM, Lyons KM, Lane TF. Wnt10b deficiency results in age-dependent loss of bone mass and progressive reduction of mesenchymal progenitor cells. J Bone Miner Res : 2138–2147, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Tamasi JA, Vasilov A, Shimizu E, Benton N, Johnson J, Bitel CL, Morrison N, Partridge NC. Monocyte chemoattractant protein-1 is a mediator of the anabolic action of parathyroid hormone on bone. J Bone Miner Res : 1975–1986, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, Zhao L, Nagy TR, Peng X, Hu J, Feng X, Van Hul W, Wan M, Cao X. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med : 757–765, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Teitelbaum SL. Bone resorption by osteoclasts. Science : 1504–1508, 2000. [DOI] [PubMed] [Google Scholar]
- 177.Valin A, Guillen C, Esbrit P. C-terminal parathyroid hormone-related protein (PTHrP) (107–139) stimulates intracellular Ca2+ through a receptor different from the type 1 PTH/PTHrP receptor in osteoblastic osteosarcoma UMR 106 cells. Endocrinology : 2752–2759, 2001. [DOI] [PubMed] [Google Scholar]
- 178.Wahl P, Wolf M. FGF23 in chronic kidney disease. Adv Exp Med Biol : 107–125, 2012. [DOI] [PubMed] [Google Scholar]
- 179.Wakisaka A, Tanaka H, Barnes J, Liang CT. Effect of locally infused IGF-I on femoral gene expression and bone turnover activity in old rats. J Bone Miner Res : 13–19, 1998. [DOI] [PubMed] [Google Scholar]
- 180.Wang K, Yamamoto H, Chin JR, Werb Z, Vu TH. Epidermal growth factor receptor-deficient mice have delayed primary endochondral ossification because of defective osteoclast recruitment. J Biol Chem : 53848–53856, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Wang Y, Nishida S, Boudignon BM, Burghardt A, Elalieh HZ, Hamilton MM, Majumdar S, Halloran BP, Clemens TL, Bikle DD. IGF-I receptor is required for the anabolic actions of parathyroid hormone on bone. J Bone Miner Res : 1329–1337, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Wang Y, Nishida S, Elalieh HZ, Long RK, Halloran BP, Bikle DD. Role of IGF-I signaling in regulating osteoclastogenesis. J Bone Miner Res : 1350–1358, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Weinstein RS. Clinical practice. Glucocorticoid-induced bone disease. N Engl J Med : 62–70, 2011. [DOI] [PubMed] [Google Scholar]
- 184.Wiren KM. Androgens and bone growth: it's location, location, location. Curr Opin Pharmacol : 626–632, 2005. [DOI] [PubMed] [Google Scholar]
- 185.Xian L, Wu X, Pang L, Lou M, Rosen CJ, Qiu T, Crane J, Frassica F, Zhang L, Rodriguez JP, Xiaofeng J, Shoshana Y, Shouhong X, Argiris E, Mei W, Xu C. Matrix IGF-1 maintains bone mass by activation of mTOR in mesenchymal stem cells. Nat Med : 1095–1101, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Xiao L, Sobue T, Esliger A, Kronenberg MS, Coffin JD, Doetschman T, Hurley MM. Disruption of the Fgf2 gene activates the adipogenic and suppresses the osteogenic program in mesenchymal marrow stromal stem cells. Bone : 360–370, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Yaccoby S, Ling W, Zhan F, Walker R, Barlogie B, Shaughnessy JD Jr. Antibody-based inhibition of DKK1 suppresses tumor-induced bone resorption and multiple myeloma growth in vivo. Blood : 2106–2111, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Yakar S, Bouxsein ML, Canalis E, Sun H, Glatt V, Gundberg C, Cohen P, Hwang D, Boisclair Y, Leroith D, Rosen CJ. The ternary IGF complex influences postnatal bone acquisition and the skeletal response to intermittent parathyroid hormone. J Endocrinol : 289–299, 2006. [DOI] [PubMed] [Google Scholar]
- 189.Yakar S, Courtland HW, Clemmons D. IGF-1 and bone: New discoveries from mouse models. J Bone Miner Res : 2543–2552, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Yakar S, Rosen CJ. From mouse to man: redefining the role of insulin-like growth factor-I in the acquisition of bone mass. Exp Biol Med (Maywood) : 245–252, 2003. [DOI] [PubMed] [Google Scholar]
- 191.Yakar S, Rosen CJ, Beamer WG, Ackert-Bicknell CL, Wu Y, Liu JL, Ooi GT, Setser J, Frystyk J, Boisclair YR, LeRoith D. Circulating levels of IGF-1 directly regulate bone growth and density. J Clin Invest : 771–781, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Yakar S, Rosen CJ, Bouxsein ML, Sun H, Mejia W, Kawashima Y, Wu Y, Emerton K, Williams V, Jepsen K, Schaffler MB, Majeska RJ, Gavrilova O, Gutierrez M, Hwang D, Pennisi P, Frystyk J, Boisclair Y, Pintar J, Jasper H, Domene H, Cohen P, Clemmons D, LeRoith D. Serum complexes of insulin-like growth factor-1 modulate skeletal integrity and carbohydrate metabolism. FASEB J : 709–719, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Yamazaki K, Suda N, Kuroda T. Immunohistochemical localization of parathyroid hormone-related protein in developing mouse Meckel's cartilage and mandible. Arch Oral Biol : 787–794, 1997. [DOI] [PubMed] [Google Scholar]
- 194.Yoshizawa T, Handa Y, Uematsu Y, Takeda S, Sekine K, Yoshihara Y, Kawakami T, Arioka K, Sato H, Uchiyama Y, Masushige S, Fukamizu A, Matsumoto T, Kato S. Mice lacking the vitamin D receptor exhibit impaired bone formation, uterine hypoplasia and growth retardation after weaning. Nat Genet : 391–396, 1997. [DOI] [PubMed] [Google Scholar]
- 195.Zhang M, Xuan S, Bouxsein ML, von Stechow D, Akeno N, Faugere MC, Malluche H, Zhao G, Rosen CJ, Efstratiadis A, Clemens TL. Osteoblast-specific knockout of the insulin-like growth factor (IGF) receptor gene reveals an essential role of IGF signaling in bone matrix mineralization. J Biol Chem : 44005–44012, 2002. [DOI] [PubMed] [Google Scholar]
- 196.Zhang X, Tamasi J, Lu X, Zhu J, Chen H, Tian X, Lee TC, Threadgill DW, Kream BE, Kang Y, Partridge NC, Qin L. Epidermal growth factor receptor plays an anabolic role in bone metabolism in vivo. J Bone Miner Res : 1022–1034, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Zhu J, Jia X, Xiao G, Kang Y, Partridge NC, Qin L. EGF-like ligands stimulate osteoclastogenesis by regulating expression of osteoclast regulatory factors by osteoblasts: implications for osteolytic bone metastases. J Biol Chem : 26656–26664, 2007. [DOI] [PubMed] [Google Scholar]
- 198.Zhu J, Shimizu E, Zhang X, Partridge NC, Qin L. EGFR signaling suppresses osteoblast differentiation and inhibits expression of master osteoblastic transcription factors Runx2 and Osterix. J Cell Biochem : 1749–1760, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]

