Abstract
Osteoimmunology is field of research dedicated to the study of the interactions between the immune system and bone. Among the cells of the immune system that regulate bone turnover and the responsiveness of bone cells to calciothropic hormones are bone marrow T lymphocytes. T cells secrete osteoclastogenic cytokines such as RANKL and TNF-α, as well as factors that stimulate bone formation, one of which is Wnt10b. In addition, T cells regulate the differentiation and life span of stromal cells and their responsiveness to parathyroid hormone (PTH) via costimulatory molecules expressed on their surface. The conditioning effect of T cells on stromal cells (SCs) is inherited by the osteoblastic and osteocytic progeny of SCs. As a result, osteoblastic cells of T cell–deficient mice have functional characteristics different from corresponding cells of T cell–replete mice. These differences include the ratio of RANKL/OPG produced in response to continuous PTH treatment, and the osteoblastogenic response to intermittent PTH treatment. This article reviews the evidence indicating that the effects of parathyroid hormone are mediated not only by osteoblasts and osteocytes but also by T cells.
Keywords: T cells, PTH, osteoblasts, osteocytes, stromal cells, bone
Introduction
Osteoimmunology is a discipline dedicated to the study of the interactions between the immune system and bone. Clinicians who have reported an association between inflammation and bone loss and increased fracture risk have long suspected the existence of important interaction between immune and bone physiology. 1-3 For example, low-grade systemic inflammation, indicated by moderately elevated serum levels of high sensitivity C-reactive protein, associates with low bone density, elevated bone resorption, and increased fracture risk.1, 2, 4 At the cellular level it is now well established that during inflammation osteoclastic bone resorption is driven by inflammatory cytokines produced by activated T cells.5 The discovery that the RANKL–RANK system plays a pivotal role in both adaptive immunity and osteoclastogenesis provided molecular evidence that has firmly established a link between the immune system and bone. The subsequent finding that RANKL produced by T cells contributes to the bone loss observed in adjuvant-induced arthritis 5 provided the first incontrovertible evidence for the capacity of some T cell lineages to cause bone loss. Initially recognized as a lineage capable of stimulating bone resorption, bone marrow (BM) T cells are known to also regulate bone formation. T cells regulate bone homeostasis via direct interactions with bone cells mediated by cell surface molecules, and by releasing cytokines and Wnt ligands. This article focuses on the role of BM T cells, osteoblasts, and osteocytes, and their interactions in the mechanism of action of PTH in bone.
T lymphocytes relevant to bone
Bone remodeling is regulated by interactions between bone cells and BM cells. Therefore, the lymphocytes critical for bone remodeling are those that home and reside in the BM. Accounting for only ~5 % of total BM cells, T cells are highly mobile and distributed among the stroma and parenchyma. 6, 7 T cells are a heterogeneous population that includes αβ and γδ T cell populations. These two families differ in the chains (αβ or γδ) forming their T cell receptor. αβ T cells are divided into naive and memory CD4+ and CD8+ T cell populations. Naive CD4+ cells differentiate into TH1, TH2, TH17, and regulatory T (Treg) effector cells. Some T cells express the cell surface marker of natural killer (NK) cells, and are thus referred to as NKT cells.
The BM is regarded as a niche for the recruitment and retention of central memory CD8+ cells.6 The BM as a lower proportion of CD4+ cells as compared to peripheral blood, and in fact the CD4+/CD8+ ratio is <1 in the BM, while is typically 2:1 in other lymphoid organs.6 Memory CD8+ cells have a higher activation state than peripheral blood T cells, and consequently secrete relatively high levels of effector cytokines. Disease state may further alter the number and phenotype of BM T cells, leading to a further exacerbation of the pathological process. For example, postmenopausal women with osteoporotic fractures have a higher proportion of TNF-α–producing CD8+ cell as compared to age-matched control women 9, 10 Some T cell lineages (e.g.,TH17 cells) have been recognized as cells capable of stimulating bone resorption, while others (e.g., Treg cells) have been shown to inhibit osteoclastogenesis; but there is increasing recognition that T cells may function as activator of bone formation, as they can secrete Wnt ligands that activate Wnt signaling in osteoblastic cells.11-13
The net effect of T cells depends on their activation state and by their specific phenotype. Activated CD4+ and CD8+ T cells promote bone loss in inflammatory diseases such as rheumatoid arthritis5 and periodontitis.14 Activated T cells also play a critical role in bone cancers15 and in post-menopausal osteoporosis.9, 10 Conversely, resting CD4+ T cells may contribute to dampening of bone resorption in vivo.16 Accordingly, T cell–deficient mice have significantly increased bone resorption and reduced bone density, as compared to controls.17 Activated conventional CD4+ and CD8+ T cells and TH17 cells regulate bone homeostasis via surface bound molecules that bind to cognate molecules expressed in osteoblasts and their stromal cell (SC) precursors18 and by releasing osteoclastogenic cytokines such as RANKL, TNF-α, IL-1, IL-6, and IL-17. Activated CD8+ T cells can also release Wnt ligands such as Wn10b11-13 that activate Wnt signaling, thus promoting osteoblastogenesis. Naive helper T cells differentiate into several lineages including TH1, TH2, and TH17 cells in response to cytokines secreted by bone cells spontaneously or in response to calicotropic hormones. By regulating T cells differentiation via their cytokines, bone cells lead to expansion of mature T cells populations that further regulate bone homeostasis 20.
The most osteoclastogenic subsets of T cells are TH17 cells.21 These cells are defined by their capacity to produce the cytokine IL-17. Therefore, CD4+ IL-17+ cells are referred to as TH17 cells. TH17 cells are constitutively present at mucosal surfaces, especially in the intestinal lamina propria,22 and are relatively frequent in the BM 23, 24 because bone is a large reservoir of TGF-β and IL-6, factors essential for TH17 cell differentiation. TH17 cells play a pivotal role in the bone loss of inflammatory conditions such as psoriasis, rheumatoid arthritis, periodontal disease, and inflammatory bowel disease (IBD).25, 26 TH17 cells potently induce osteoclastogenesis by secreting IL-17, RANKL, TNF-α, IL-1, and IL-6, along with low levels of IFN-γ.27-29 IL-17 stimulates the release of RANKL by osteoblasts and osteocytes21, 30 and potentiates the osteoclastogenic activity of RANKL by upregulating RANK.31 IL-17 provides an important connection between T cells and osteocytes, as this T cell cytokine regulates osteocytic RANKL production,30 a key effect of PTH on osteocytes.32, 33 TH17 cells have been linked to postmenopausal osteoporosis because the differentiation of TH17 cells is induced by ovariectomy (ovx),34 while the bone loss induced by ovx is prevented by silencing of IL-17R 35 and treatment with anti-IL-17.36 Importantly, elevated levels of IL-17 have been found in postmenopausal women with osteoporosis.37-39
The most bone-sparing population of T cells are Treg cells, a suppressive population of CD4+ T cells defined by the expression of the transcription factor FoxP3 and the ability to block conventional T cell proliferation and production of effector cytokines.40 Treg cells form when naive CD4+ cells are exposed to antigen in the presence of TGF-β. Defects in Treg cell numbers and/or activity have been implicated in several chronic inflammatory diseases. As a result, there has been a recent explosion in research investigating the potential to manipulate Treg cells for clinical purposes 41-45. It is now clear that Treg cells regulate osteoclast (OC) formation 46-49 and prevent ovx-induced bone loss.50 Treg cells blunt bone resorption 47, 51 through the secretion of IL-4, IL-10, and TGF-β.49 Estrogen increases the relative number of Treg cells.52 It is interesting to note that the cytokines produced by Treg cells to repress effector T cells also possess a strong anti- osteoclastogenic activity. Intravital microscopy studies have disclosed that Treg cells are not randomly distributed in the BM but rather reside in close proximity with endosteal bone surfaces and osteoclasts (OB).53 Osteoclasts selectively recruit and activate CD8+ T cells,54, 55 which express CD25 and FoxP3 and therefore are defined as osteoclast-induced regulatory CD8 T cells.56 Moreover, a recent report has disclosed that although RANKL directly stimulates osteoclasts to resorb bone, at lower doses this cytokine also controls osteoclast ability to induce regulatory T cells, an important negative feedback loop. Attesting to relevance, ovariectomized mice treated with low-dose RANKL exhibit CD8+ Treg cells that suppress bone resorption, decrease the levels of inflammatory/osteoclastogenic cytokines, and stimulate bone formation. These studies suggest the existence of a novel regulatory loop, whereby osteoclasts and the osteoclast-inducing factor RANKL induce Treg cells, and then the Treg cells blunt osteoclastic bone resorption.57
T cells make use of several mechanisms to regulate bone cells. First, they are armed with surface costimulatory molecules that bind their counter receptors expressed in bone cells. For example, T cells express RANKL and CD40L, which activate the cognate receptors RANK and CD40 in osteoclast precursors and osteoblastic cells, respectively.18, 20, 58, 59 CD40L, also known as CD154, exerts its effects by binding to CD40 60 and several integrins.61-64 CD40 is expressed on antigen presenting cells,65 SCs, and osteoblasts.66 CD40–CD40L is crucial for T cell activation and several functions of the immune system. It promotes macrophage activation and differentiation, antibody isotype switching, and the adequate organization of immunologic memory in B cells.67 Binding to the integrins αIIbβ3,61, 62 Mac-1,63 and α5β1, which are widely expressed in the BM, is known to account for additional inflammatory effects of CD40L 64.
CD40L has been linked to post-natal skeletal maturation because children affected by X-linked hyper-IgM syndrome––a condition in which CD40L production is impaired due to a mutation of the CD40L gene––have low bone density.68 Two mechanisms have been identified to link the CD40L–CD40 to skeletal maturation. First, activation of CD40 signaling in B cells by T cell expressed CD40L promotes production of the anti-osteoclastogenic factor OPG by B cells,16 thereby decreasing bone resorption. In addition, activation of CD40 signaling in SCs by T cell–expressed CD40L provides proliferative and survival cues to SCs both in vitro and in vivo.58, 66 CD40L also increases the commitment of SCs to the osteoblastic lineage.58, 69 At the same time, activation of CD40 signaling in osteoblastic cells increases their osteoclastogenic activity,58, 69 which is defined as the capacity of osteoblastic cells to induce and support osteoclast formation in vitro. Attesting to its relevance, CD40L is required for ovariectomy and continuous PTH treatment––a model of primary hyperparathyroidism––to expand SCs and their osteoblastic progeny and to increase the osteoclastogenic activity of SCs 18, 58. Underscoring the complexity of this regulatory system, T cells also suppress osteoclast formation by inducing CD80/86 signaling in osteoclast precursors by inducing the IDO/tryptophan pathway 8.
Skeletal effects of PTH
Parathyroid hormone is an important endocrine regulator of calcium and phosphorus concentrations in extracellular fluid. Physiologic levels of circulating PTH are essential for maintaining serum and urinary calcium levels within their normal range. Chronic excessive PTH production is a cause of skeletal and extra skeletal disease. Secondary hyperparathyroidism has been implicated in the pathogenesis of senile osteoporosis,70 while primary hyperparathyroidism (PHPT), is associated with accelerated bone loss,71 osteopenia,72-74 and increased bone turnover,73 an independent risk factor for fractures. Furthermore, PHPT is a cause of extra-skeletal manifestations stemming from increased bone resorption such as hypercalcemia, recurrent nephrolithiasis, renal failure, peptic ulcers and mental changes.72 Primary and secondary hyperparathyroidism are mimicked by continuous PTH (cPTH) infusion. However when injected daily, a regimen known as intermittent PTH (iPTH) treatment, the hormone markedly stimulates trabecular and cortical bone formation. Although this bone forming activity is antagonized, in part, by a stimulation of bone resorption, the net effect of iPTH is an improvement in bone microarchitecture and increased strength.75-77 As a result, intermittent treatment with the 1–34 fragment of PTH decreases the risk of fractures in humans, and is a U.S. Food and Drug Administration approved treatment modality for postmenopausal osteoporosis.78, 79 iPTH treatment is also under investigation as an agent for accelerating fracture repair and diminish the risk of non-unions.
Both cPTH and iPTH increase bone turnover in trabecular and cortical bone, as evidenced by elevations in histomorphometric and biochemical markers of resorption and formation,80-82 whereas PHPT and cPTH treatment cause cortical bone loss by enhancing endosteal resorption through stimulation of OC formation, activity and life span.72, 82, 83 Severe chronic elevations of PTH levels may also lead to trabecular bone loss,72, 82 although PHPT and cPTH treatment often induce a modest increase in cancellous bone.73, 74, 80, 84 In contrast, iPTH treatment markedly increases trabecular bone volume due to a preponderant stimulation of trabecular bone formation, and causes a small loss of cortical bone.75, 78 PTH promotes bone formation by increasing the number of osteoblasts85-87 through multiple effects, including activation of quiescent lining cells,88 increased OB proliferation89, 90 and differentiation, 89, 91, 92 and attenuation of pre-OB and OB apoptosis.93-96
PTH exerts its biological activities by binding to the PPR receptor (also known as PTH-1R), which is expressed on SCs, osteoblasts and osteocytes97-99 but also T cells13 and macrophages.100 SCs and osteoblasts were the first targets of PTH to be identified and earlier consensus developed that the catabolic effect of cPTH is mostly mediated by enhanced production of RANKL and decreased production of OPG by SCs and osteoblasts.76, 101, 102 More recent studies in mice with deletion and/or overexpression of PPR and RANKL in osteocytes32, 33, 99, 103 lead to the recognition that osteocytes represent essential targets of PTH in bone, and that increased production of RANKL by osteocytes plays an important role in cPTH induced bone loss.32, 33 However, some reports have ascribed a key role to OB produced RANKL.104 osteocytes have also emerged as a critical target for the anabolic effects of PTH because transgenic mice expressing a constitutively active PTH receptor exclusively in osteocytes exhibit increased bone mass and bone remodeling, as well as reduced expression of the osteocyte-derived Wnt antagonist sclerostin, increased Wnt signaling, and decreased OB apoptosis.105 Direct regulation of sclerostin production by osteocytes is regarded as a particularly relevant role in the anabolic activity of PTH.105
One of the major effects of PTH is to activate Wnt signaling in all osteoblastic cells, including osteocytes. Activation of Wnt signaling induces OB proliferation106 and differentiation, 107 prevents pre-OB and OB apoptosis,95 and augments OB production of OPG.108 Wnt proteins initiate a canonical signaling cascade by binding to receptors of the Frizzled family together with coreceptors, members of the low-density lipoprotein receptor-related protein (LRP) family, LRP5 and LRP6, which results in the stabilization of cytosolic β-catenin. A nuclear complex of β-catenin and the T cell factor/lymphoid enhancer factor (TCF/LEF) family of transcription factors then interacts with DNA to regulate the transcription of Wnt target genes.109 PTH is a canonical Wnt signaling agonist that increases β-catenin levels in osteoblastic cells.110 PTH, once bound to PPR, is also capable of forming a complex with LRP6 which results in LRP6 signaling and β-catenin activation.111 Thus, PTH activates Wnt signaling in osteoblastic cells through both Wnt ligand–dependent and Wnt ligand–independent mechanisms.
The capacity of PTH to suppress the production of sclerostin,112-114 the finding that serum levels of sclerostin are inversely correlated with PTH levels in healthy women,115 and reports that women treated with teriparatide have decreased serum levels of sclerostin116 have led to the hypothesis that repression of the sclerostin gene (SOST) and the resulting inhibition of sclerostin production are a key mechanism of action of iPTH.117 Studies in Sost transgenic and global Sost−/− mice revealed that sclerostin plays an important, yet non-exclusive, role in iPTH-induced anabolism.118-120 The existence of a sclerostin-independent effect of iPTH was supported by the finding that iPTH equally elevates bone formation and resorption in WT and in Sost transgenic mice.118 In addition, iPTH induced a significant increase in trabecular thickness and mineral apposition rate in Sost−/− mice, thus demonstrating that iPTH stimulates bone formation also independently of sclerostin suppression.118 Initial studies could not provide an answer to the question of whether iPTH increases trabecular bone volume in the absence of sclerostin, as no increase in bone mass was detected in the distal femur of both growing WT control and Sost−/− mice in one study,118 while full anabolic activity of iPTH in the trabecular bone of skeletally-mature Sost−/− mice was reported in another.120 Uncertainty concerning the relevance of sclerostin-independent modalities of action of iPTH is the confounding effect of the altered baseline bone density that is characteristic of mice either lacking or overexpressing sclerostin.118-120 The fact that iPTH blunts but does not completely block sclerostin production further limits the usefulness of Sost−/− mice as a tool to investigate the mechanism of action of iPTH
PTH also regulates Dickkopf-1, a soluble LRP5 and LRP6 signaling inhibitor,110 and Sfrp-4, a factor that binds Wnt proteins and thereby antagonizes both canonical and non-canonical Wnt signaling.121 Uncertainty remains with regard to the identity and the source of Wnt ligands that activate Wnt signaling in response to PTH treatment.
BM macrophages are additional lineages involved in the mechanism of action of PTH. Depletion of macrophage precursors results in osteopenia and blunted iPTH anabolic activity. 100 By contrast, depletion of mature phagocytic macrophages potentiates iPTH-dependent anabolism by activating efferocytosis, a process that stimulates other BM cells to secrete Wn10b, Wnt3a, and TGF-β.100 Moreover, iPTH stimulates macrophages to secrete TGF-β through an indirect effect mediated by sIL-6R, which is secreted by osteoblasts in response to iPTH.122 Although macrophages express the PTH receptor PPR,100 the available data suggest that BMMs play a permissive role in the anabolic activity of iPTH. Another lineage known to express PPR are dendritic cells (DCs). 123 However, DCs have not been implicated in the mechanism of action of PTH in bone.
Role of T cells in primary hyperparathyroidism and the catabolic activity of continuous PTH treatment
T lymphocytes express functional PPR,13 respond to PTH,124 and stimulate OB differentiation 125. These observations prompted more in depth investigations on the role of T cells as mediators of the pro-resorptive effect of cPTH treatment. These studies revealed that infusion of cPTH that mimics hyperparathyroidism fails to induce OC formation, bone resorption and cortical bone loss in mice lacking T cells.58 By contrast, cPTH equally stimulated bone formation in T cell–replete and T cell–1deficient mice. Interestingly, transplantation of tumors producing PTH and/or PTHrP in nude mice, a strain of T cell–deficient mice, stimulates bone resorption in spite of the absence of T cells in the host.126-128 The apparent discrepancy between these reports and those in which mice were treated with cPTH is explained by the higher levels of circulating PTH/PTHrP attained by transplanting PTH/PTHrP-producing tumors, as compared to those obtained by infusing cPTH at 80. Adding further support to this hypothesis are reports demonstrating that transplantation of adenomatous and hyperplastic parathyroid tissues in nude mice, in a fashion that elevates serum PTH to 80–240 ng/L, fails to induce hypercalcemia,129, 130 while parathyroid transplants that elevate serum PTH to ~1000 ng/L induce hypercalcemia in nude mice.131
T cells exert complex effects that are relevant for the effects of PTH in bone. The most upstream effect of cPTH is that of stimulating the production of TNF-α by both CD4+ and CD8+ T cells.132 Since CD8+ cells are more abundant in the BM than CD4+ cells, most of the TNF-α produced in the BM in response to cPTH originates from CD8+ cells.132 Attesting to the relevance of T cell produced TNF-α, cPTH fails to induce bone loss and stimulate bone resorption in mice specifically lacking T cell TNF-α production.132 PTH induces T cell production of TNF-α via direct activation of PPR signaling in T cells.132 Conditional silencing of the PTH receptor PPR in T cells blunts the stimulation of bone resorption induced by cPTH without affecting bone formation, thus blocking cortical bone loss and converting the effects of cPTH in trabecular bone from catabolic to anabolic.132 These findings demonstrate the critical relevance of direct PPR signaling in T cells. TNF-α stimulates osteoclast formation and activity via multiple mechanisms, which include increased production of RANKL by all osteoblastic cells including osteocytes. An important early effect of TNF-α is that of upregulating the expression of CD40 in SCs and osteoblasts, which increase their response to T cell expressed CD40L. Activation of CD40 signaling in osteoblastic cells increases the sensitivity of SCs and osteoblasts to cPTH, leading to a smaller suppression of OPG secretion in response to PTH and a resulting increase in the RANKL/OPG ratio. It should be noted that cPTH stimulates bone cells and immune cells to release growth factors and cytokines. Among them are TGF-β, IL-6, and TNF-α.122, 132-134 TGF-β and IL-6 direct the differentiation of naive CD4+ cells into TH17 cells. 26, 135, 136 Studies with agents neutralizing TNF-α have implicated TNF-α in the generation of TH17 cells in rodents and humans.137-139 Recent data indeed suggest that cPTH-induce TH17 cell differentiation, and that IL-17A produced by TH17 cells, may act as an upstream cytokine that plays a pivotal role in the bone loss induced by cPTH and primary hyperparathyroidism.30 A fertile area of future investigation will be to determine the role of IL-17 in the bone loss caused by cPTH and PHPT.
Role of T cells in the anabolic activity of intermittent PTH treatment
Studies conducted in T cell deficient mice revealed that mice lacking T cells exhibit a blunted increase in bone formation and trabecular bone volume in response to iPTH 13.
The importance of T cells for the bone anabolic activity of iPTH is shown in Figure 1, which depicts longitudinal sections of the femurs from WT and TCRβ-deficient (Tcrb−/−) mice treated with vehicle and iPTH. Tcrb−/− mice are characterized by a complete deficiency of aβ T cells. iPTH treatment for 4 weeks leads to the appearance of cuboidal lining osteoblasts in WT but not in Tcrb−/− mice. Since cuboidal osteoblasts are actively engaged in bone formation, the images suggest that iPTH is not able to increase bone formation in Tcrb−/− mice.
Figure 1.
Trichrome stained longitudinal sections of femurs from WT and Tcrb−/− mice treated with daily injections of vehicle or human PTH (80 microgam/Kg/day) for 4 weeks. Lining osteoblasts acquire the typical morphology of bone forming osteoblasts (cuboidal appearance) in WT mice but not in Tcrb−/− mice. A representative image is shown.
Furthermore, adoptive transfer of T cells into T cell deficient mice restored a normal bone anabolic response to iPTH. T cells augment the capacity of iPTH to improve architecture in trabecular but not in cortical bone. Although the reason of this selectivity is unknown, a lack of access of T cells to cortical surfaces is not a likely explanation, as T cells reach endosteal and periosteal bone surface through blood vessels and recirculate in and out of the BM.140 With regard to the mechanism by which T cells potentiate the bone anabolic activity of iPTH, it is now clear that in the absence of T cells, iPTH is unable to increase the commitment of SCs to the osteoblastic lineage, induce OB proliferation and differentiation, and mitigate OB apoptosis. All of these actions of PTH were found to hinge on the capacity of T cells to activate Wnt signaling in osteoblastic cells.13 Although it is well established that Wnt activation is a key mechanism by which iPTH expands the osteoblastic pool, little information is available on the nature and the source of the Wnt ligand required to activate Wnt signaling in SCs and osteoblasts. BM CD8+ T cells produce large amounts of the osteogenic Wnt ligand Wnt10b.13 The relevance of CD8+ cells was demonstrated by the inability of iPTH to promote bone anabolism in major histocompatibility complex (MHC) class I–deficient mice, a strain that lacks CD8+ cells.13 Additional studies revealed that iPTH does not improve bone architecture in T cell–deficient mice reconstituted with CD4+ cells, while it does so in mice adoptively transferred with CD8+ cells.13 The pivotal role of T cell produced Wnt10b was revealed by the hampered effect of iPTH on bone volume in global Wnt10b−/− mice 141 and T cell–deficient mice reconstituted with T cells from Wnt10b−/− mice.13 Together, the data indicate that CD8+ T cells potentiates the anabolic activity of PTH by providing Wnt10b, which is a critical Wnt ligand required for activating Wnt signaling in osteoblastic cells. Therefore, in the absence of CD8+ cells, stimulation of osteoblastic cells by PTH is not sufficient to elicit maximal Wnt activation due to the lack of a critical Wnt ligand.
Importantly, a recent report in human has shown that treatment with teriparatide, a form of iPTH treatment, increases the BM levels of Wnt10b.142 That study has also shown that T cells are the main source of Wnt10b in humans treated with teriparatide.142 By contrast, patients affected by in primary hyperparathyroidism do not exhibit increased Wnt10b expression.142
Additional interactions between T cells and SCs may contribute to the anabolic activity of iPTH. An important mediator of T cell–SC interaction is the T cell costimulatory ligand CD40L. 143 Attesting to the relevance of CD40L in vivo, silencing of CD40L blocks the effects of iPTH on SC proliferation, differentiation, and life span, resulting in blunted anabolic activity of iPTH on trabecular bone.
Together, the data suggest a requirement for a dual regulatory interaction between T cells and SCs. According to this model, silencing of either Wnt10b or CD40L is sufficient to blunt the responsiveness of SCs and their osteoblastic progeny to iPTH.
The residual bone anabolic activity of PTH observed in T cell–deficient mice is presumably due to ligand independent activation of LRP6,111 and suppressed production of sclerostin.105, 112, 113 Indeed, it has recently been reported that mice treated with anti-sclerostin antibody maintain a partial anabolic response to iPTH. The sclerostin-independent activity of iPTH has shown to be completely due to increased production of Wnt10b by T cells.144
In summary, the available data are consistent with a complex modality of action of iPTH that include suppression of sclerostin production and increased T cell production of Wnt10b (Fig. 2). In conditions of normal baseline bone turnover and bone mass and partial sclerostin blockade that mimic the repressive activity of the hormone on sclerostin production, iPTH stimulates osteoblastogenesis, OB life span, bone density, and trabecular bone volume independently of sclerostin through a Wnt10b-mediated mechanism.
Figure 2.
Schematic representation of the mechanism of action of iPTH. In baseline conditions, T cells produce low levels of Wnt10b, whereas osteocytes secrete high levels of sclerostin. The presence of a low Wnt10b/sclerostin ratio prevents the activation of Wnt signaling in osteoblasts, leading to low osteoblastogenesis and high OB apoptosis. Treatment with iPTH increases the T cell production of Wnt10b and blunts the osteocytic secretion of sclerostin, thus increasing the Wnt10b/sclerostin ratio. Wnt10b is then capable of activating Wnt signaling in osteoblasts, leading to increased osteoblastogenesis and decreased OB apoptosis. Reproduced with permission from Ref. 144.
Osteoblasts, osteocytes, and T cells are key targets of PTH
As discussed above, robust data have been published by many laboratories showing that silencing of PPR signaling in osteoblasts, osteocytes, or T cells results in the partial or total blockade of the bone anabolic and catabolic effects of PTH. These findings may sound surprising and perhaps contradictory if one envisions PTH acting using a parallel circuit regulatory modality. In this model, PTH targets osteoblasts, osteocytes, and T cells independently, and interactions between immune cells and bone cells do not play a significant role. This model forces investigators to focus on the effects of PTH on one cell population, for example on the capacity of the hormone to stimulate the osteocytic production of RANKL, and to underestimate the contributions of the effects of PTH on another lineage, for example the capacity to induce T cell production of TNF-α.
However, the fact that silencing of PPR signaling in one lineage (e.g., T cells) induces the same effects as silencing of PPR signaling in another (e.g., osteocytes) is in keeping with a serial circuit regulatory mode (Fig. 3). This model finds it justification on the fact that BM T cells provide cell surface signals and secrete cytokines that direct the differentiation of SCs toward osteoblasts characterized by a high sensitivity to PTH. Conversely, SCs that differentiate in a microenvironment devoid of T cells acquire a permanent low sensitivity to PTH, which is transmitted to their osteoblastic progeny (Fig. 4). Since osteoblasts differentiate into osteocytes, low responsive osteocyte will arise from low responsive osteoblasts. Thus, by acting on SCs and early OB precursors, T cells have the capacity to set the responsiveness of all osteoblastic cells to PTH, including osteocytes. In addition, cytokines secreted by T cells, such as TNF-α and IL-17, target mature osteoblasts and osteocytes, regulating their production of RANKL.30 We thus hypothesize the existence of a serial, rather than parallel, connection between T cells, osteoblasts, and osteocytes, with reciprocal, bidirectional regulatory interactions. T cells are upstream targets of PTH, while osteoblasts and osteocytes are downstream targets. Therefore, T cells, osteoblasts, and osteocytes are all critical targets of PTH.
Figure 3.
Diagrammatic representation of published data demonstrating that silencing of PPR signaling in either T cells, osteoblasts, or osteocytes leads to either decreased or absent responses to cPTH and iPTH.32, 33, 99, 103, 132, 155 This type of response is consistent with a serial circuit regulatory mechanism.
Figure 4.
T cells lead to the differentiation of stromal cells (SCs) into PTH responsive osteoblasts (osteoblasts) and osteocytes (osteocytes). CD40L, a surface molecules expressed on T cells activates CD40 signaling in SCs, that selects and expands a population of SCs highly responsive to PTH. As SCs differentiate into osteoblasts and osteocytes, T cell–derived signaling conditions the entire osteoblastic progeny. (A) SCs from T cell–replete mice differentiate into osteoblasts and osteocytes highly sensitive to PTH. (B) SCs, osteoblasts, and osteocytes from T cell–deficient mice exhibit low sensitivity to PTH.
Conclusions
An impressive amount of work published in the last 10 years has led to the recognition that T cells play an unexpected role in the regulation of bone resorption and bone formation through a variety of mechanisms. Studies in humans have corroborated observations in mice and shown the relevance of T cells for bone health in humans. Most of the evidence in humans has accrued in studies on the pathogenesis of postmenopausal osteoporosis. For example, evidence begins to emerge in favor of a role of T cell–produced TNF-α in postmenopausal bone loss in women,9, 10 and that in humans estrogen deficiency expands RANKL-expressing T and B cells.145, 146 Moreover, a role for IL-1 and TNF-α in humans is supported by reports that menopause increases the levels of these factors,147-151 while treatment with inhibitors of IL-1 and TNF-α prevents the increase in bone resorption induced by estrogen deficiency.152
Evidence has also emerged that T cells may contribute to the bone anabolic activity of teriparatide treatment in humans. These observations represent the first attempt to translate in humans the large body of evidence pointing to a role of T cells in the mechanism of action of PTH in the mouse. Preliminary reports show that hyperparathyroidism may elevate IL-17 production in humans, suggesting that human confirmation may be found for the studies on cPTH and T cells conducted in the mouse.
The most intriguing aspect of PTH biology remains the opposite response of the skeleton to continuous, versus intermittent, exposure to PTH. Several hypotheses have been proposed to explain the divergent response to cPTH and iPTH. Among them is a differential effect of cPTH and iPTH on OB life span, an hypothesis based on the observation that iPTH attenuates OB apoptosis93-96 while cPTH does not.94 However, this explanation does not account for the differences in bone resorption elicited by iPTH and cPTH. In vitro studies have led to the alternative hypothesis that the bone response to PTH reflects the intermittent or continuous activation of PPR in bone cells.153, 154 However, this hypothesis does not explain why transgenic mice expressing a constitutively active PPR in osteoblasts or osteocytes exhibit a dramatic increase in trabecular bone formation97, 105 that resembles trabecular bone formation induced by iPTH. The capacity of T cells to secrete TNF-α and Wnt10 in response to cPTH and iPTH introduces a novel layer of complexity, but also offers new opportunities to fully understand the mechanism of action of PTH in bone.
Acknowledgements
This paper was supported by grants from the National Institutes of Health (AR54625, DK007298, and RR028009).
Footnotes
Conflicts of interest The author declares no conflicts of interest
REFERENCES
- 1.Ding C, et al. Circulating levels of inflammatory markers predict change in bone mineral density and resorption in older adults: a longitudinal study. J Clin Endocrinol Metab. 2008;93:1952–1958. doi: 10.1210/jc.2007-2325. [DOI] [PubMed] [Google Scholar]
- 2.Pasco JA, et al. High-sensitivity C-reactive protein and fracture risk in elderly women. JAMA : the journal of the American Medical Association. 2006;296:1353–1355. doi: 10.1001/jama.296.11.1353. [DOI] [PubMed] [Google Scholar]
- 3.Eriksson AL, et al. High-sensitivity CRP is an independent risk factor for all fractures and vertebral fractures in elderly men: the MrOS Sweden study. J Bone Miner Res. 2014;29:418–423. doi: 10.1002/jbmr.2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Schett G, et al. High-sensitivity C-reactive protein and risk of nontraumatic fractures in the Bruneck study. Archives of internal medicine. 2006;166:2495–2501. doi: 10.1001/archinte.166.22.2495. [DOI] [PubMed] [Google Scholar]
- 5.Kong YY, et al. Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature. 1999;402:304–309. doi: 10.1038/46303. [DOI] [PubMed] [Google Scholar]
- 6.Di Rosa F, Pabst R. The bone marrow: a nest for migratory memory T cells. Trends in immunology. 2005;26:360–366. doi: 10.1016/j.it.2005.04.011. [DOI] [PubMed] [Google Scholar]
- 7.Zhao E, et al. Bone marrow and the control of immunity. Cellular & molecular immunology. 2012;9:11–19. doi: 10.1038/cmi.2011.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bozec A, et al. T cell costimulation molecules CD80/86 inhibit osteoclast differentiation by inducing the IDO/tryptophan pathway. Sci Transl Med. 2014;6:235ra260. doi: 10.1126/scitranslmed.3007764. [DOI] [PubMed] [Google Scholar]
- 9.D’Amelio P, et al. Estrogen deficiency increases osteoclastogenesis up-regulating T cells activity: a key mechanism in osteoporosis. Bone. 2008;43:92–100. doi: 10.1016/j.bone.2008.02.017. [DOI] [PubMed] [Google Scholar]
- 10.Adeel S, et al. Bone loss in surgically ovariectomized premenopausal women is associated with T lymphocyte activation and thymic hypertrophy. J Investig Med. 2013;61:1178–1183. doi: 10.231/JIM.0000000000000016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hardiman G, et al. The mouse Wnt-10B gene isolated from helper T cells is widely expressed and a possible oncogene in BR6 mouse mammary tumorigenesis. Gene. 1996;172:199–205. doi: 10.1016/0378-1119(96)00109-6. [DOI] [PubMed] [Google Scholar]
- 12.Ouji Y, et al. Wnt-10b secreted from lymphocytes promotes differentiation of skin epithelial cells. Biochem Biophys Res Commun. 2006;342:1063–1069. doi: 10.1016/j.bbrc.2006.02.028. [DOI] [PubMed] [Google Scholar]
- 13.Terauchi M, et al. T lymphocytes amplify the anabolic activity of parathyroid hormone through Wnt10b signaling. Cell metabolism. 2009;10:229–240. doi: 10.1016/j.cmet.2009.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Teng YT, et al. Functional human T-cell immunity and osteoprotegerin ligand control alveolar bone destruction in periodontal infection. J Clin Invest. 2000;106:59–67. doi: 10.1172/jci10763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang K, et al. CD8+ T cells regulate bone tumor burden independent of osteoclast resorption. Cancer Res. 2011;71:4799–4808. doi: 10.1158/0008-5472.CAN-10-3922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Li Y, et al. B cells and T cells are critical for the preservation of bone homeostasis and attainment of peak bone mass in vivo. Blood. 2007;109:3839–3848. doi: 10.1182/blood-2006-07-037994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Toraldo G, et al. IL-7 induces bone loss in vivo by induction of receptor activator of nuclear factor kappa B ligand and tumor necrosis factor alpha from T cells. Proc Natl Acad Sci U S A. 2003;100:125–130. doi: 10.1073/pnas.0136772100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Li JY, et al. Ovariectomy disregulates osteoblast and osteoclast formation through the T-cell receptor CD40 ligand. Proc Natl Acad Sci U S A. 2011;108:768–773. doi: 10.1073/pnas.1013492108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pacifici R. T cells: critical bone regulators in health and disease. Bone. 2010;47:461–471. doi: 10.1016/j.bone.2010.04.611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pacifici R. Osteoimmunology and its implications for transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2013;13:2245–2254. doi: 10.1111/ajt.12380. [DOI] [PubMed] [Google Scholar]
- 21.Sato K, et al. Th17 functions as an osteoclastogenic helper T cell subset that links T cell activation and bone destruction. J Exp Med. 2006;203:2673–2682. doi: 10.1084/jem.20061775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ivanov II, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–1133. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
- 23.Kordasti SY, et al. IL-17-producing CD4(+) T cells, pro-inflammatory cytokines and apoptosis are increased in low risk myelodysplastic syndrome. Br J Haematol. 2009;145:64–72. doi: 10.1111/j.1365-2141.2009.07593.x. [DOI] [PubMed] [Google Scholar]
- 24.Kappel LW, et al. IL-17 contributes to CD4-mediated graft-versus-host disease. Blood. 2009;113:945–952. doi: 10.1182/blood-2008-08-172155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med. 2009;361:888–898. doi: 10.1056/NEJMra0707449. [DOI] [PubMed] [Google Scholar]
- 26.Martinez GJ, et al. Regulation and function of proinflammatory TH17 cells. Ann N Y Acad Sci. 2008;1143:188–211. doi: 10.1196/annals.1443.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Komatsu N, Takayanagi H. Autoimmune arthritis: the interface between the immune system and joints. Advances in immunology. 2012;115:45–71. doi: 10.1016/B978-0-12-394299-9.00002-3. [DOI] [PubMed] [Google Scholar]
- 28.Waisman A. T helper cell populations: as flexible as the skin? European journal of immunology. 2011;41:2539–2543. doi: 10.1002/eji.201141938. [DOI] [PubMed] [Google Scholar]
- 29.Jovanovic DV, et al. IL-17 stimulates the production and expression of proinflammatory cytokines, IL-beta and TNF-alpha, by human macrophages. J Immunol. 1998;160:3513–3521. [PubMed] [Google Scholar]
- 30.JY L, et al. IL-17A is increased in humans with primary hyperparathyroidism and mediates PTH-induced bone loss in mice. Cell metabolism. 2015 doi: 10.1016/j.cmet.2015.09.012. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Adamopoulos IE, et al. Interleukin-17A upregulates receptor activator of NF-kappaB on osteoclast precursors. Arthritis Res Ther. 2010;12:R29. doi: 10.1186/ar2936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Xiong J, et al. Osteocyte-derived RANKL is a critical mediator of the increased bone resorption caused by dietary calcium deficiency. Bone. 2014;66C:146–154. doi: 10.1016/j.bone.2014.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Saini V, et al. Parathyroid hormone (PTH)/PTH-related peptide type 1 receptor (PPR) signaling in osteocytes regulates anabolic and catabolic skeletal responses to PTH. The Journal of biological chemistry. 2013;288:20122–20134. doi: 10.1074/jbc.M112.441360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tyagi AM, et al. Estrogen deficiency induces the differentiation of IL-17 secreting Th17 cells: a new candidate in the pathogenesis of osteoporosis. PLoS ONE. 2012;7:e44552. doi: 10.1371/journal.pone.0044552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.DeSelm CJ, et al. IL-17 mediates estrogen-deficient osteoporosis in an Act1-dependent manner. Journal of cellular biochemistry. 2012;113:2895–2902. doi: 10.1002/jcb.24165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tyagi AM, et al. Enhanced immunoprotective effects by anti-IL-17 antibody translates to improved skeletal parameters under estrogen deficiency compared with anti-RANKL and anti-TNF-alpha antibodies. J Bone Miner Res. 2014;29:1981–1992. doi: 10.1002/jbmr.2228. [DOI] [PubMed] [Google Scholar]
- 37.Molnar I, Bohaty I, Somogyine-Vari E. IL-17A-mediated sRANK ligand elevation involved in postmenopausal osteoporosis. Osteoporos Int. 2014;25:783–786. doi: 10.1007/s00198-013-2548-6. [DOI] [PubMed] [Google Scholar]
- 38.Zhang J, et al. Changes of serum cytokines-related Th1/Th2/Th17 concentration in patients with postmenopausal osteoporosis. Gynecological endocrinology : the official journal of the International Society of Gynecological Endocrinology1-8. 2014 doi: 10.3109/09513590.2014.975683. [DOI] [PubMed] [Google Scholar]
- 39.Molnar I, Bohaty I, Somogyine-Vari E. High prevalence of increased interleukin-17A serum levels in postmenopausal estrogen deficiency. Menopause. 2014;21:749–752. doi: 10.1097/GME.0000000000000125. [DOI] [PubMed] [Google Scholar]
- 40.Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30:636–645. doi: 10.1016/j.immuni.2009.04.010. [DOI] [PubMed] [Google Scholar]
- 41.Trzonkowski P, et al. First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127- T regulatory cells. Clin Immunol. 2009;133:22–26. doi: 10.1016/j.clim.2009.06.001. [DOI] [PubMed] [Google Scholar]
- 42.Brunstein CG, et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood. 2011;117:1061–1070. doi: 10.1182/blood-2010-07-293795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Di Ianni M, et al. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood. 2011;117:3921–3928. doi: 10.1182/blood-2010-10-311894. [DOI] [PubMed] [Google Scholar]
- 44.Marek-Trzonkowska N, et al. Administration of CD4+CD25highCD127- regulatory T cells preserves beta-cell function in type 1 diabetes in children. Diabetes care. 2012;35:1817–1820. doi: 10.2337/dc12-0038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Feng F, et al. Display of human proinsulin on the Bacillus subtilis spore surface for oral administration. Current microbiology. 2013;67:1–8. doi: 10.1007/s00284-013-0325-6. [DOI] [PubMed] [Google Scholar]
- 46.Zaiss MM, et al. Treg cells suppress osteoclast formation: a new link between the immune system and bone. Arthritis Rheum. 2007;56:4104–4112. doi: 10.1002/art.23138. [DOI] [PubMed] [Google Scholar]
- 47.Kim YG, et al. Human CD4+CD25+ regulatory T cells inhibit the differentiation of osteoclasts from peripheral blood mononuclear cells. Biochem Biophys Res Commun. 2007;357:1046–1052. doi: 10.1016/j.bbrc.2007.04.042. [DOI] [PubMed] [Google Scholar]
- 48.Kelchtermans H, et al. Activated CD4+CD25+ regulatory T cells inhibit osteoclastogenesis and collagen-induced arthritis. Ann Rheum Dis. 2009;68:744–750. doi: 10.1136/ard.2007.086066. [DOI] [PubMed] [Google Scholar]
- 49.Luo CY, et al. Estrogen enhances the functions of CD4(+)CD25(+)Foxp3(+) regulatory T cells that suppress osteoclast differentiation and bone resorption in vitro. Cellular & molecular immunology. 2011;8:50–58. doi: 10.1038/cmi.2010.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zaiss MM, et al. Increased bone density and resistance to ovariectomy-induced bone loss in FoxP3-transgenic mice based on impaired osteoclast differentiation. Arthritis and rheumatism. 2010;62:2328–2338. doi: 10.1002/art.27535. [DOI] [PubMed] [Google Scholar]
- 51.Yuan FL, et al. Regulatory T cells as a potent target for controlling bone loss. Biochemical and biophysical research communications. 2010;402:173–176. doi: 10.1016/j.bbrc.2010.09.120. [DOI] [PubMed] [Google Scholar]
- 52.Tai P, et al. Induction of regulatory T cells by physiological level estrogen. Journal of cellular physiology. 2008;214:456–464. doi: 10.1002/jcp.21221. [DOI] [PubMed] [Google Scholar]
- 53.Fujisaki J, et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature. 2011;474:216–219. doi: 10.1038/nature10160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kiesel J, et al. Systems level analysis of osteoclastogenesis reveals intrinsic and extrinsic regulatory interactions. Dev Dyn. 2007;236:2181–2197. doi: 10.1002/dvdy.21206. [DOI] [PubMed] [Google Scholar]
- 55.Li H, et al. Cross talk between the bone and immune systems: osteoclasts function as antigen-presenting cells and activate CD4+ and CD8+ T cells. Blood. 2010;116:210–217. doi: 10.1182/blood-2009-11-255026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Buchwald ZS, et al. Osteoclast activated FoxP3+ CD8+ T-cells suppress bone resorption in vitro. PLoS ONE. 2012;7:e38199. doi: 10.1371/journal.pone.0038199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Buchwald ZS, et al. A Bone Anabolic Effect of RANKL in a Murine Model of Osteoporosis Mediated Through FoxP3(+) CD8 T Cells. J Bone Miner Res. 2015;30:1508–1522. doi: 10.1002/jbmr.2472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gao Y, et al. T cells potentiate PTH-induced cortical bone loss through CD40L signaling. Cell metabolism. 2008;8:132–145. doi: 10.1016/j.cmet.2008.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Di Rosa F. T-lymphocyte interaction with stromal, bone and hematopoietic cells in the bone marrow. Immunol Cell Biol. 2009;87:20–29. doi: 10.1038/icb.2008.84. [DOI] [PubMed] [Google Scholar]
- 60.Quezada SA, et al. CD40/CD154 interactions at the interface of tolerance and immunity. Annual review of immunology. 2004;22:307–328. doi: 10.1146/annurev.immunol.22.012703.104533. [DOI] [PubMed] [Google Scholar]
- 61.Andre P, et al. CD40L stabilizes arterial thrombi by a beta3 integrin--dependent mechanism. Nat Med. 2002;8:247–252. doi: 10.1038/nm0302-247. [DOI] [PubMed] [Google Scholar]
- 62.Prasad KS, et al. Soluble CD40 ligand induces beta3 integrin tyrosine phosphorylation and triggers platelet activation by outside-in signaling. Proc Natl Acad Sci U S A. 2003;100:12367–12371. doi: 10.1073/pnas.2032886100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zirlik A, et al. CD40 ligand mediates inflammation independently of CD40 by interaction with Mac-1. Circulation. 2007;115:1571–1580. doi: 10.1161/CIRCULATIONAHA.106.683201. [DOI] [PubMed] [Google Scholar]
- 64.Leveille C, et al. CD40 ligand binds to alpha5beta1 integrin and triggers cell signaling. The Journal of biological chemistry. 2007;282:5143–5151. doi: 10.1074/jbc.M608342200. [DOI] [PubMed] [Google Scholar]
- 65.Grammer AC, Lipsky PE. CD40-mediated regulation of immune responses by TRAF-dependent and TRAF-independent signaling mechanisms. Advances in immunology. 2000;76:61–178. doi: 10.1016/s0065-2776(01)76019-1. [DOI] [PubMed] [Google Scholar]
- 66.Ahuja SS, et al. CD40 ligand blocks apoptosis induced by tumor necrosis factor alpha, glucocorticoids, and etoposide in osteoblasts and the osteocyte-like cell line murine long bone osteocyte-Y4. Endocrinology. 2003;144:1761–1769. doi: 10.1210/en.2002-221136. [DOI] [PubMed] [Google Scholar]
- 67.Grewal IS, Flavell RA. The role of CD40 ligand in costimulation and T-cell activation. Immunol Rev. 1996;153:85–106. doi: 10.1111/j.1600-065x.1996.tb00921.x. [DOI] [PubMed] [Google Scholar]
- 68.Lopez-Granados E, et al. Osteopenia in X-linked hyper-IgM syndrome reveals a regulatory role for CD40 ligand in osteoclastogenesis. Proc Natl Acad Sci U S A. 2007;104:5056–5061. doi: 10.1073/pnas.0605715104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Li JY, et al. Ovariectomy expands murine short-term hemopoietic stem cell function through T cell expressed CD40L and Wnt10B. Blood. 2013;122:2346–2357. doi: 10.1182/blood-2013-03-487801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Riggs BL, Melton LJ. Medical progress: involutional osteoporosis. N. Eng. J. Med. 1986;314:1676–1684. doi: 10.1056/NEJM198606263142605. [DOI] [PubMed] [Google Scholar]
- 71.Grey AB, et al. Accelerated bone loss in post-menopausal women with mild primary hyperparathyroidism. Clin Endocrinol (Oxf) 1996;44:697–702. doi: 10.1046/j.1365-2265.1996.744565.x. [DOI] [PubMed] [Google Scholar]
- 72.Potts J. Primary hyperparathyroidism. In: Avioli LV, Krane S, editors. Metabolic Bone Diseases. Vol. 1. Academic Press; San Diego: 1998. pp. 411–442. [Google Scholar]
- 73.Parisien M, et al. The parathyroids. Basic and clinical concepts. Academic Press; San Diego: 2001. Histomorphometric analysis of bone in primary hyperparathyroidism; pp. 423–436. [Google Scholar]
- 74.Silverberg SJ, et al. Skeletal disease in primary hyperparathyroidism. J Bone Miner Res. 1989;4:283–291. doi: 10.1002/jbmr.5650040302. [DOI] [PubMed] [Google Scholar]
- 75.Dempster DW, et al. Anabolic actions of parathyroid hormone on bone. Endocr Rev. 1993;14:690–709. doi: 10.1210/edrv-14-6-690. [DOI] [PubMed] [Google Scholar]
- 76.Qin L, Raggatt LJ, Partridge NC. Parathyroid hormone: a double-edged sword for bone metabolism. Trends Endocrinol Metab. 2004;15:60–65. doi: 10.1016/j.tem.2004.01.006. [DOI] [PubMed] [Google Scholar]
- 77.Zaidi M. Skeletal remodeling in health and disease. Nat Med. 2007;13:791–801. doi: 10.1038/nm1593. [DOI] [PubMed] [Google Scholar]
- 78.Neer RM, et al. Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001;344:1434–1441. doi: 10.1056/NEJM200105103441904. [DOI] [PubMed] [Google Scholar]
- 79.Canalis E, Giustina A, Bilezikian JP. Mechanisms of anabolic therapies for osteoporosis. N Engl J Med. 2007;357:905–916. doi: 10.1056/NEJMra067395. [DOI] [PubMed] [Google Scholar]
- 80.Zhou H, et al. Continuous parathyroid hormone and estrogen administration increases vertebral cancellous bone volume and cortical width in the estrogen-deficient rat. J Bone Miner Res. 2001;16:1300–1307. doi: 10.1359/jbmr.2001.16.7.1300. [DOI] [PubMed] [Google Scholar]
- 81.Dempster DW, et al. On the mechanism of cancellous bone preservation in postmenopausal women with mild primary hyperparathyroidism. J Clin Endocrinol Metab. 1999;84:1562–1566. doi: 10.1210/jcem.84.5.5652. [DOI] [PubMed] [Google Scholar]
- 82.Iida-Klein A, et al. Short-term continuous infusion of human parathyroid hormone 1-34 fragment is catabolic with decreased trabecular connectivity density accompanied by hypercalcemia in C57BL/J6 mice. J Endocrinol. 2005;186:549–557. doi: 10.1677/joe.1.06270. [DOI] [PubMed] [Google Scholar]
- 83.Lotinun S, et al. Continuous parathyroid hormone induces cortical porosity in the rat: effects on bone turnover and mechanical properties. J Bone Miner Res. 2004;19:1165–1171. doi: 10.1359/JBMR.040404. [DOI] [PubMed] [Google Scholar]
- 84.Hock JM, Gera I. Effects of continuous and intermittent administration and inhibition of resorption on the anabolic response of bone to parathyroid hormone. J Bone Miner Res. 1992;7:65–72. doi: 10.1002/jbmr.5650070110. [DOI] [PubMed] [Google Scholar]
- 85.Lindsay R, et al. A novel tetracycline labeling schedule for longitudinal evaluation of the short-term effects of anabolic therapy with a single iliac crest bone biopsy: early actions of teriparatide. J Bone Miner Res. 2006;21:366–373. doi: 10.1359/JBMR.051109. [DOI] [PubMed] [Google Scholar]
- 86.Ma YL, et al. Teriparatide increases bone formation in modeling and remodeling osteons and enhances IGF-II immunoreactivity in postmenopausal women with osteoporosis. J Bone Miner Res. 2006;21:855–864. doi: 10.1359/jbmr.060314. [DOI] [PubMed] [Google Scholar]
- 87.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. 1997;138:4607–4612. doi: 10.1210/endo.138.11.5505. [DOI] [PubMed] [Google Scholar]
- 88.Dobnig H, Turner RT. Evidence that intermittent treatment with parathyroid hormone increases bone formation in adult rats by activation of bone lining cells. Endocrinology. 1995;136:3632–3638. doi: 10.1210/endo.136.8.7628403. [DOI] [PubMed] [Google Scholar]
- 89.Nishida S, et al. Increased bone formation by intermittent parathyroid hormone administration is due to the stimulation of proliferation and differentiation of osteoprogenitor cells in bone marrow. Bone. 1994;15:717–723. doi: 10.1016/8756-3282(94)90322-0. [DOI] [PubMed] [Google Scholar]
- 90.Pettway GJ, et al. Parathyroid hormone mediates bone growth through the regulation of osteoblast proliferation and differentiation. Bone. 2007 doi: 10.1016/j.bone.2007.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Meng XW, et al. Temporal expression of the anabolic action of PTH in cancellous bone of ovariectomized rats. J Bone Miner Res. 1996;11:421–429. doi: 10.1002/jbmr.5650110402. [DOI] [PubMed] [Google Scholar]
- 92.Schmidt IU, Dobnig H, Turner RT. Intermittent parathyroid hormone treatment increases osteoblast number, steady state messenger ribonucleic acid levels for osteocalcin, and bone formation in tibial metaphysis of hypophysectomized female rats. Endocrinology. 1995;136:5127–5134. doi: 10.1210/endo.136.11.7588250. [DOI] [PubMed] [Google Scholar]
- 93.Jilka RL, et al. Increased bone formation by prevention of osteoblast apoptosis with parathyroid hormone. J Clin invest. 1999;104:439–446. doi: 10.1172/JCI6610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bellido T, et al. Proteasomal degradation of Runx2 shortens parathyroid hormone-induced anti-apoptotic signaling in osteoblasts. A putative explanation for why intermittent administration is needed for bone anabolism. The Journal of biological chemistry. 2003;278:50259–50272. doi: 10.1074/jbc.M307444200. [DOI] [PubMed] [Google Scholar]
- 95.Almeida M, et al. Wnt proteins prevent apoptosis of both uncommitted osteoblast progenitors and differentiated osteoblasts by beta-catenin-dependent and -independent signaling cascades involving Src/ERK and phosphatidylinositol 3-kinase/AKT. The Journal of biological chemistry. 2005;280:41342–41351. doi: 10.1074/jbc.M502168200. [DOI] [PubMed] [Google Scholar]
- 96.Tobimatsu T, et al. Parathyroid hormone increases beta-catenin levels through Smad3 in mouse osteoblastic cells. Endocrinology. 2006;147:2583–2590. doi: 10.1210/en.2005-1627. [DOI] [PubMed] [Google Scholar]
- 97.Calvi LM, et al. Activated parathyroid hormone/parathyroid hormone-related protein receptor in osteoblastic cells differentially affects cortical and trabecular bone. J Clin Invest. 2001;107:277–286. doi: 10.1172/JCI11296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lanske B, et al. Ablation of the PTHrP gene or the PTH/PTHrP receptor gene leads to distinct abnormalities in bone development. J Clin Invest. 1999;104:399–407. doi: 10.1172/JCI6629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Powell WF, Jr., et al. Targeted ablation of the PTH/PTHrP receptor in osteocytes impairs bone structure and homeostatic calcemic responses. J Endocrinol. 2011;209:21–32. doi: 10.1530/JOE-10-0308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Cho SW, et al. Osteal macrophages support physiologic skeletal remodeling and anabolic actions of parathyroid hormone in bone. Proc Natl Acad Sci U S A. 2014 doi: 10.1073/pnas.1315153111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Weir EC, et al. Colony stimulating factor-1 plays a role in osteoclast formation and function in bone resorption induced by parathyroid hormone and parathyroid hormone-related protein. J Bone Miner Res. 1996;11:1474–1481. doi: 10.1002/jbmr.5650111014. [DOI] [PubMed] [Google Scholar]
- 102.Ma YL, et al. Catabolic effects of continuous human PTH (1--38) in vivo is associated with sustained stimulation of RANKL and inhibition of osteoprotegerin and gene-associated bone formation. Endocrinology. 2001;142:4047–4054. doi: 10.1210/endo.142.9.8356. [DOI] [PubMed] [Google Scholar]
- 103.Ben-awadh AN, et al. Parathyroid hormone receptor signaling induces bone resorption in the adult skeleton by directly regulating the RANKL gene in osteocytes. Endocrinology. 2014;155:2797–2809. doi: 10.1210/en.2014-1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Fumoto T, et al. Physiological functions of osteoblast lineage and T cell-derived RANKL in bone homeostasis. J Bone Miner Res. 2014;29:830–842. doi: 10.1002/jbmr.2096. [DOI] [PubMed] [Google Scholar]
- 105.O’Brien CA, et al. Control of bone mass and remodeling by PTH receptor signaling in osteocytes. PLoS ONE. 2008;3:e2942. doi: 10.1371/journal.pone.0002942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kato M, et al. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–314. doi: 10.1083/jcb.200201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Jilka RL. Molecular and cellular mechanisms of the anabolic effect of intermittent PTH. Bone. 2007;40:1434–1446. doi: 10.1016/j.bone.2007.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Glass DA, 2nd, et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Developmental cell. 2005;8:751–764. doi: 10.1016/j.devcel.2005.02.017. [DOI] [PubMed] [Google Scholar]
- 109.Behrens J, et al. Functional interaction of beta-catenin with the transcription factor LEF-1. Nature. 1996;382:638–642. doi: 10.1038/382638a0. [DOI] [PubMed] [Google Scholar]
- 110.Kulkarni NH, et al. Effects of parathyroid hormone on Wnt signaling pathway in bone. Journal of cellular biochemistry. 2005;95:1178–1190. doi: 10.1002/jcb.20506. [DOI] [PubMed] [Google Scholar]
- 111.Wan M, et al. Parathyroid hormone signaling through low-density lipoprotein-related protein 6. Genes Dev. 2008;22:2968–2979. doi: 10.1101/gad.1702708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Keller H, Kneissel M. SOST is a target gene for PTH in bone. Bone. 2005;37:148–158. doi: 10.1016/j.bone.2005.03.018. [DOI] [PubMed] [Google Scholar]
- 113.Bellido T, et al. Chronic elevation of parathyroid hormone in mice reduces expression of sclerostin by osteocytes: a novel mechanism for hormonal control of osteoblastogenesis. Endocrinology. 2005;146:4577–4583. doi: 10.1210/en.2005-0239. [DOI] [PubMed] [Google Scholar]
- 114.Silvestrini G, et al. Effects of intermittent parathyroid hormone (PTH) administration on SOST mRNA and protein in rat bone. J Mol Histol. 2007;38:261–269. doi: 10.1007/s10735-007-9096-3. [DOI] [PubMed] [Google Scholar]
- 115.Mirza FS, et al. Serum sclerostin levels negatively correlate with parathyroid hormone levels and free estrogen index in postmenopausal women. J Clin Endocrinol Metab. 2010;95:1991–1997. doi: 10.1210/jc.2009-2283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Drake MT, et al. Effects of parathyroid hormone treatment on circulating sclerostin levels in postmenopausal women. J Clin Endocrinol Metab. 2010;95:5056–5062. doi: 10.1210/jc.2010-0720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kramer I, et al. Does osteocytic SOST suppression mediate PTH bone anabolism? Trends Endocrinol Metab. 2010;21:237–244. doi: 10.1016/j.tem.2009.12.002. [DOI] [PubMed] [Google Scholar]
- 118.Kramer I, et al. Parathyroid hormone (PTH)-induced bone gain is blunted in SOST overexpressing and deficient mice. J Bone Miner Res. 2010;25:178–189. doi: 10.1359/jbmr.090730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sims NA. Building bone with a SOST-PTH partnership. J Bone Miner Res. 2010;25:175–177. doi: 10.1002/jbmr.53. [DOI] [PubMed] [Google Scholar]
- 120.Robling AG, et al. Anabolic and Catabolic Regimens of Human Parathyroid Hormone 1-34 Elicit Bone- and Envelope-Specific Attenuation of Skeletal Effects in Sost-Deficient Mice. Endocrinology. 2011 doi: 10.1210/en.2011-0049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Qin L, et al. Gene expression profiles and transcription factors involved in parathyroid hormone signaling in osteoblasts revealed by microarray and bioinformatics. The Journal of biological chemistry. 2003;278:19723–19731. doi: 10.1074/jbc.M212226200. [DOI] [PubMed] [Google Scholar]
- 122.Cho SW, et al. The soluble interleukin-6 receptor is a mediator of hematopoietic and skeletal actions of parathyroid hormone. The Journal of biological chemistry. 2013 doi: 10.1074/jbc.M112.393363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gessi M, et al. Parathyroid hormone-related peptide (hPTHrP) and parathyroid hormone-related peptide receptor type 1 (PTHR1) expression in human thymus. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2005;53:955–962. doi: 10.1369/jhc.4A6548.2005. [DOI] [PubMed] [Google Scholar]
- 124.Stojceva-Taneva O, et al. Parathyroid hormone increases cytosolic calcium of thymocytes. Nephron. 1993;64:592–599. doi: 10.1159/000187406. [DOI] [PubMed] [Google Scholar]
- 125.Rifas L. T-cell cytokine induction of BMP-2 regulates human mesenchymal stromal cell differentiation and mineralization. Journal of cellular biochemistry. 2006;98:706–714. doi: 10.1002/jcb.20933. [DOI] [PubMed] [Google Scholar]
- 126.Guise TA, et al. Chinese hamster ovarian cells transfected with human parathyroid hormone-related protein cDNA cause hypercalcemia in nude mice. Lab Invest. 1992;67:477–485. [PubMed] [Google Scholar]
- 127.Mehdizadeh S, et al. Bone resorption and circulating PTH-like bioactivity in an animal model of hypercalcaemia of malignancy. Biochem Biophys Res Commun. 1989;161:1166–1171. doi: 10.1016/0006-291x(89)91364-8. [DOI] [PubMed] [Google Scholar]
- 128.Uy HL, et al. Effects of parathyroid hormone (PTH)-related protein and PTH on osteoclasts and osteoclast precursors in vivo. Endocrinology. 1995;136:3207–3212. doi: 10.1210/endo.136.8.7628353. [DOI] [PubMed] [Google Scholar]
- 129.Hory BG, et al. Absence of response to human parathyroid hormone in athymic mice grafted with human parathyroid adenoma, hyperplasia or parathyroid cells maintained in culture. J Endocrinol Invest. 2000;23:273–279. doi: 10.1007/BF03343723. [DOI] [PubMed] [Google Scholar]
- 130.Strieth S, et al. A new animal model to assess angiogenesis and endocrine function of parathyroid heterografts in vivo. Transplantation. 2005;79:392–400. doi: 10.1097/01.tp.0000151633.92173.75. [DOI] [PubMed] [Google Scholar]
- 131.Schachter P, et al. Function in athymic nude mice of parathyroid heterografts from patients with primary hyperparathyroidism and secondary hyperparathyroidism. Surgery. 1990;108:1040–1046. [PubMed] [Google Scholar]
- 132.Tawfeek H, et al. Disruption of PTH Receptor 1 in T Cells Protects against PTH-Induced Bone Loss. PLoS ONE. 2010;5:e12290. doi: 10.1371/journal.pone.0012290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Lowik CWGM, et al. Parathyroid hormone (PTH) and PTH-like protein (PLP) stimulate IL-6 production by osteogenic cells: a possible role of interleukin-6 in osteoclastogensis. Biochem.Biophys.Res.Commun. 1989;162:1546–1552. doi: 10.1016/0006-291x(89)90851-6. [DOI] [PubMed] [Google Scholar]
- 134.Koh AJ, et al. An irradiation-altered bone marrow microenvironment impacts anabolic actions of PTH. Endocrinology. 2011;152:4525–4536. doi: 10.1210/en.2011-1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bettelli E, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- 136.Basu R, Hatton RD, Weaver CT. The Th17 family: flexibility follows function. Immunol Rev. 2013;252:89–103. doi: 10.1111/imr.12035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nakae S, et al. Mast cell-derived TNF can promote Th17 cell-dependent neutrophil recruitment in ovalbumin-challenged OTII mice. Blood. 2007;109:3640–3648. doi: 10.1182/blood-2006-09-046128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chen DY, et al. Increasing levels of circulating Th17 cells and interleukin-17 in rheumatoid arthritis patients with an inadequate response to anti-TNF-alpha therapy. Arthritis Res Ther. 2011;13:R126. doi: 10.1186/ar3431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sugita S, et al. Inhibition of Th17 differentiation by anti-TNF-alpha therapy in uveitis patients with Behcet’s disease. Arthritis Res Ther. 2012;14:R99. doi: 10.1186/ar3824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Di Rosa F. T-lymphocyte interaction with stromal, bone and hematopoietic cells in the bone marrow. Immunol Cell Biol. 2008;87:20–29. doi: 10.1038/icb.2008.84. [DOI] [PubMed] [Google Scholar]
- 141.Bedi B, et al. Silencing of parathyroid hormone (PTH) receptor 1 in T cells blunts the bone anabolic activity of PTH. Proc Natl Acad Sci U S A. 2012 doi: 10.1073/pnas.1120735109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Bianchi ML, et al. Improving adherence to and persistence with oral therapy of osteoporosis. Osteoporos Int. 2015;26:1629–1638. doi: 10.1007/s00198-015-3038-9. [DOI] [PubMed] [Google Scholar]
- 143.Grewal IS, Flavell RA. CD40 and CD154 in cell-mediated immunity. Annual review of immunology. 1998;16:111–135. doi: 10.1146/annurev.immunol.16.1.111. [DOI] [PubMed] [Google Scholar]
- 144.Li JY, et al. The sclerostin-independent bone anabolic activity of intermittent PTH treatment is mediated by T-cell-produced Wnt10b. J Bone Miner Res. 2014;29:43–54. doi: 10.1002/jbmr.2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Eghbali-Fatourechi G, et al. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest. 2003;111:1221–1230. doi: 10.1172/JCI17215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Taxel P, et al. Estradiol rapidly inhibits osteoclastogenesis and RANKL expression in bone marrow cultures in postmenopausal women: a pilot study. Osteoporos Int. 2008;19:193–199. doi: 10.1007/s00198-007-0452-7. [DOI] [PubMed] [Google Scholar]
- 147.Pacifici R, et al. Ovarian steroid treatment blocks a postmenopausal increase in blood monocyte interleukin 1 release. Proc Natl Acad Sci U S A. 1989;86:2398–2402. doi: 10.1073/pnas.86.7.2398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Pacifici R, et al. Effect of surgical menopause and estrogen replacement on cytokine release from human blood mononuclear cells. Proc Natl Acad Sci U S A. 1991;88:5134–5138. doi: 10.1073/pnas.88.12.5134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Cohen-Solal ME, et al. Peripheral monocyte culture supernatants of menopausal women can induce bone resorption: involvement of cytokines. J Clin Endocrinol Metab. 1993;77:1648–1653. doi: 10.1210/jcem.77.6.8263153. [DOI] [PubMed] [Google Scholar]
- 150.Cohen-Solal ME, et al. Bone resorption at the femoral neck is dependent on local factors in nonosteoporotic late postmenopausal women: an in vitro-in vivo study. J Bone Miner Res. 1995;10:307–314. doi: 10.1002/jbmr.5650100219. [DOI] [PubMed] [Google Scholar]
- 151.Bernard-Poenaru O, et al. Bone-resorbing cytokines from peripheral blood mononuclear cells after hormone replacement therapy: a longitudinal study. Osteoporos Int. 2001;12:769–776. doi: 10.1007/s001980170054. [DOI] [PubMed] [Google Scholar]
- 152.Charatcharoenwitthaya N, et al. Effect of Blockade of Tumor Necrosis Factor-alpha and Interleukin-1 Action on Bone Resorption in Early Postmenopausal Women. J Bone Miner Res. 2007 doi: 10.1359/jbmr.070207. [DOI] [PubMed] [Google Scholar]
- 153.Ishizuya T, et al. Parathyroid hormone exerts disparate effects on osteoblast differentiation depending on exposure time in rat osteoblastic cells. J Clin Invest. 1997;99:2961–2970. doi: 10.1172/JCI119491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Locklin RM, et al. Mediators of the biphasic responses of bone to intermittent and continuously administered parathyroid hormone. Journal of cellular biochemistry. 2003;89:180–190. doi: 10.1002/jcb.10490. [DOI] [PubMed] [Google Scholar]
- 155.Bedi B, et al. Silencing of parathyroid hormone (PTH) receptor 1 in T cells blunts the bone anabolic activity of PTH. Proc Natl Acad Sci U S A. 2012;109:E725–733. doi: 10.1073/pnas.1120735109. [DOI] [PMC free article] [PubMed] [Google Scholar]




