Open access

Mechanism of Glucocorticoid-Induced Osteoporosis: An Update

Written By

Xing-Ming Shi, Norman Chutkan, Mark W. Hamrick and Carlos M. Isales

Submitted: 23 May 2012 Published: 28 November 2012

DOI: 10.5772/53978

From the Edited Volume

Glucocorticoids - New Recognition of Our Familiar Friend

Edited by Xiaoxiao Qian

Chapter metrics overview

3,532 Chapter Downloads

View Full Metrics

1. Introduction

Synthetic oral steroids were initially developed in the 1940-1950’s. Although their use was initially limited by their high cost, as they became more affordable and began to be used for treatment of a wide variety of conditions, side effects associated with their use became much more prevalent. In fact, glucocorticoid-induced osteoporosis is now the most common secondary cause of osteoporosis. Until relatively recently, the mechanism of action of these drugs and the mechanisms involved in the development of side effects such as osteoporosis and the higher incidence of bone fractures was not known. Although steroids are widely viewed as mainly catabolic for bone a distinction needs to be made between physiologic and pharmacologic doses of steroids. Recent evidence demonstrates that steroids can clearly be anabolic for bone. In this chapter we review recent findings and mechanisms of glucocorticoid action on bone and some of the clinical consequences of pharmacologic doses of these compounds on bone.

Advertisement

2. Glucocorticoids and osteoporosis

Glucocorticoids are among the most potent anti-inflammatory and immunosuppressive agents and are key therapeutic agents for the management of chronic inflammatory diseases, including rheumatic diseases [1-4], pulmonary disease [5;6], asthma [7-11] and post transplantation immunotherapy [12]. However, long-term glucocorticoid therapy (>3 months) causes bone loss resulting in osteoporosis (glucocorticoid-induced osteoporosis or GIOP) [3;4;13-15], a severe-side effect that occurs in 30 – 50% of patients [16-18]. The incidence of GIOP is indiscriminate of race, age and gender [19;20]. Children, as young as 4 years of age, and adolescents who are on glucocorticoid therapy for various pediatric disorders, including asthma [20-22], juvenile rheumatoid arthritis [23;24], Crohn's disease [25], systemic lupus erythematosus [26;27], and inflammatory bowel disease [28;29] have been reported to endure significant bone density decrease. There is no clearly defined threshold for safe use of glucocorticoids. In practice, a dose equal to or greater than 5mg/day of prednisone is considered as low, and 10mg/day or more is high. The severity of bone loss in GIOP is both time- and dose-dependent. GIOP occurs in two phases: a rapid, early phase in which bone mineral density is reduced, within the first 5 to 7 months of therapy, possibly as a result of excessive bone resorption, and a slower, progressive phase in which bone mineral density declines because of impaired bone formation [30]. Bone loss continues as long as treatment is maintained.

Advertisement

3. Glucocorticoid mechanism of action as anti-inflammatory and immunosuppressant drugs

Glucocorticoids exert their actions via intracellular glucocorticoid receptors (GRs) [31;32]. The GR belongs to the ligand-regulated nuclear receptor superfamily [33]. Like other members in this superfamily, GR contains three major functional domains: a N-terminal activation domain required for transcriptional activation and association with basal transcription factors; a central DNA-binding domain (DBD) consisting of two highly conserved zinc finger regions that are critical for dimerization, DNA binding, transcriptional activation and repression; and a C-terminal ligand-binding domain (LBD) that serves as the binding site for glucocorticoids, chaperone proteins, and coactivators [34;35]. In the absence of ligand, GR is predominantly retained in the cytoplasm as an inactive multi-protein complex consisting of heat shock protein (hsp90) and a number of other proteins, including the immunophilins. The binding of glucocorticoid triggers a conformational change in the GR and leads to dissociation of the multi-protein complex and exposure of a nuclear localization sequence resulting in its nuclear translocation. Once in the nucleus, GR, in the form of a homodimer, binds to a palindromic glucocorticoid-response element (GRE) in the target gene promoter and activates transcription (e.g., of the tyrosine amino transferase gene), or it can bind to a negative GRE (nGRE) to repress transcription (e.g., of the osteocalcin gene) [36].

Glucocorticoids suppress the expression of a panel of inflammatory-relevant genes including cytokines [interleukins (IL) and tumor necrosis factors (TNF-α,β], chemokines (Regulated upon Activation Normal T-cell Expressed and Secreted or RANTES, Macrophage Inflammatory Protein-1-alpha or MIP-1α, Monocyte Chemotactic Protein or MCP-1, -3, and -4], inflammatory enzymes (COX-2, iNOS), and adhesion molecules (Intercellular Adhesion Molecule 1 or ICAM-1, E-selectin) that play a key role in the recruitment of inflammatory cells to the inflammation sites [37-39]. However, most of these genes do not have negative GREs in their promoter regions, and therefore, they are not directly regulated by the binding of GRs to such regulatory elements. These genes do contain NF-κB- and/or AP-1-binding sites and are activated through these sites by NF-κB and/or AP-1 in response to stimuli (cytokines). Thus, one mechanism by which glucocorticoids could regulate transcription would be modulation of NF-κB or AP-1 DNA-binding activity. In 1990, three independent groups found cross-talk between GR and AP-1 [40-42]. In these studies, it was found that activated GR can interact with c-Jun/AP-1 and that the formation of a GR-c-Jun complex prevents c-Jun/AP-1 DNA-binding, resulting in the inhibition of gene expression. Later, it was found that the activated GR can associate with the p65 subunit of NF-κB and inhibit gene activation mediated by NF-κB [43;44]. These findings led to the establishment of the protein-protein interaction model.

In 1995, it was found that glucocorticoids induce the expression of a cytoplasmic inhibitor of NF-κB, the IκB-α [45;46]. These studies led to the establishment of a second model, the IκB-α upregulation model. This model proposes that glucocorticoids induce the expression of IκB-α and that the newly synthesized IκB-α sequesters the p65 subunit of the NF-κB in the cytoplasm and thereby inhibits NF-κB nuclear functions. However, this mechanism has been challenged by a number of studies. It has now been established that the effect of glucocorticoids on IκB-α expression, and subsequently NF-κB nuclear translocation, is cell-type specific. In some cell types glucocorticoid inhibition of proinflammatory stimuli-induced p65 nuclear translocation is coupled with the induction of IκB-α [45-48]. In other cell types, however, these two events are uncoupled [49;50]. Moreover, a GR mutant that does not enhance IκB-α expression, is still able to repress NF-κB activity [51].

Advertisement

4. Glucocorticoid effects on bone cells

Glucocorticoids have both anabolic and catabolic effects on bone. However, the outcome of glucocorticoid therapy is a net loss of bone [4;52;53]. Corticosteroid 11β-hydroxysteroid dehydrogenase 2 [11β-HSD2] is an enzyme that oxidizes the active form of glucocorticoid cortisol to the inactive metabolite cortisone, thus the levels of expression and activity of this enzyme is critical for glucocorticoid signaling. In vivo studies show that bone-specific transgenic overexpression of 11β-HSD2, under the control of type I collagen promoter, impairs osteoblast differentiation and bone acquisition [54-56]. These studies demonstrate that the endogenous glucocorticoid signaling is essential for normal skeletal development. However, glucocorticoid in excess such as patients with Cushing’s syndrome [22] or the patients on glucocorticoid therapy rapidly lose bone mass resulting in osteoporosis. The direct effects of glucocorticoids on bone cells are illustrated in Figure 1.

Advertisement

5. Glucocorticoid effects on bone marrow Mesenchymal Stem Cells (MSCs)

Bone marrow MSCs are multipotent cells that can give rise to several distinct cell lineages, including osteoblasts, adipocytes, and chondrocytes [57-60]. Patients on glucocorticoid therapy not only lose bone but also accumulate large amounts of marrow fat (fatty marrow), indicating that glucocorticoid has altered lineage commitment of MSC to adipocytes at the expense of osteoblasts because these two pathways have a reciprocal relationship [61-64]. Thus, one possible mechanism by which glucocorticoids alter MSC fate determination is through the induction of the master adipogenic regulator peroxisome proliferator-activated receptor gamma (PPARγ) [65;66], which is transcriptionally activated by the CCAAT/enhancer binding protein (C/EBP) family transcription factors in response to glucocorticoid [67-70] (Figure 2). Indeed, Weinstein and colleagues showed that administration of glucocorticoids to mice reduces the numbers of osteoprogenitor cells [71]. This could be achieved through induction of PPARγ since under the same condition bone marrow adipogenesis is enhanced [72], and that a reduction in PPARγ dosage (haploinsufficiency) in mice results in reduced adipogenesis and enhanced osteogenesis from bone marrow progenitors [73].

Figure 1.

Glucocorticoids bind to the glucocorticoid receptor (GR) and affect mesenchymal stem cell (MSC), osteoblast (OB), osteoclast (OC) and osteocyte (Ocyte) function. The net result is decreased bone formation and increased bone resorption. ↑ increase; ↓decrease.

Figure 2.

Glucocorticoid reduces the number of osteoprogenitors from MSC by promoting adipogenic differentiation pathway. Glucocorticoid induces the expression of C/EBP family transcription factors that directly activate the transcription of PPARγ, the master regulator of adipogenesis, and shifts the lineage commitment of MSCs to adipocyte pathway, thus reducing the number of osteoprogenitor cells.

Advertisement

6. Glucocorticoid effects on osteoblasts and osteocytes

It has been known for decades that glucocorticoid inhibits bone formation [52;74], but only recently have we realized that glucocorticoids directly target bone cells. By administering a high dose of prednisolone to mice, Weinstein and colleagues found that glucocorticoid induces the death of mature osteoblasts and osteocytes [71;75]. In the same study, the authors also showed that the same is true in bone biopsy samples obtained from patients with glucocorticoid-induced osteoporosis. These results were further strengthened in a transgenic mouse model, in which the glucocorticoid signaling is disrupted by overexpression of 11β-HSD2 specifically in osteoblasts. The study showed that the 11β-HSD2 transgenic mice are protected from glucocorticoid-induced osteoblasts and osteocytes apoptosis and suppression of bone formation [76]. These studies demonstrate that glucocorticoids cause bone loss by restricting the supply of bone building cells, the osteoblasts, and by interfering with the communication network within bone environment via osteocyte death. The osteocytes are the mechanosensory cells that detect and send signals for bone formation in response to damages caused by mechanical loading and unloading [77;78].

Advertisement

7. Glucocorticoid effects on osteoclasts

Osteoclasts are bone resorbing cells and play a key role in the maintenance of bone homeostasis through bone remodeling. In patients, glucocorticoid-induced osteoporosis features a rapid early phase increase in bone resorption, followed by a slow progressive decrease in bone formation [52]. Earlier studies showed that glucocorticoids stimulate osteoclast differentiation and increase their activity [72;79;80]. It is now recognized that glucocorticoids increase the longevity of osteoclasts but may inhibit their bone resorptive activity [81;82]. Moreover, a recent study suggests that glucocorticoids do not inhibit, but modify osteoclast resorptive behavior, making osteoclasts erode bone surfaces over long distances without interruption [83].

Advertisement

8. Glucocorticoid-induced Leucine Zipper (GILZ): A new glucocorticoid anti-inflammatory effect mediator

The protein-protein interaction and the IκB-α upregulation models described earlier in this chapter were established prior to the discovery of a glucocorticoid-inducible protein named glucocorticoid-induced leucine zipper (GILZ), which was identified in 1997 [84]. GILZ is a member of the leucine zipper protein family [84;85] and belongs to the transforming growth factor-beta (TGF-β)-stimulated clone-22 (TSC-22d3) family of transcription factors [86;87]. Members of this family of proteins contain three distinct domains; an N-terminal domain containing a TSC box (N-Ter), a middle leucine zipper domain (LZ), and a C-terminal poly-proline rich domain (PRR).

Unlike IκB-α, which is induced by glucocorticoids in certain cell types [49;50;88], GILZ is induced by glucocorticoids virtually in all cell types examined so far, including bone marrow mesenchymal stem cells, osteoblasts, adipocytes, macrophages and epithelial cells [89]. In vitro studies show that overexpression of GILZ protects T-cells from apoptosis induced by anti-CD3 monoclonal antibody, but not other apoptosis-inducing agents such as dexamethasone, ultraviolet irradiation, starvation, or triggered by cross-linked anti-Fas mAb [84]. T-cell-specific transgenic overexpression of GILZ results in thymocyte apoptosis ex vivo, possibly through down-regulation of Bcl-xL [90]. The in vitro actions of GILZ have been shown to be mediated through direct protein-protein interactions between GILZ and NF-κB, and between GILZ and AP-1 [86;91;92]. The interaction between GILZ and NF-κB blocks NF-κB nuclear translocation and DNA-binding, and the interaction with AP-1 inhibits the binding of AP-1 to its DNA elements [91;92]. GILZ also interacts directly with the mitogen-activated protein kinase (MAPK) family members, Ras and Raf-1, resulting in inhibition of Raf-1 phosphorylation and subsequently, inhibition of MEK/ERK-1/2 phosphorylation and AP-1-dependent transcription [86;93]. Moreover, GILZ can deactivate macrophages [94], inhibit proinflammatory cytokine-induced inflammatory enzymes such as cyclooxygenase-2 [95], inhibit IL-2/IL-2 receptor and IL-5 expression [91;96], and stimulate the production of anti-inflammatory IL-10 by immature dendritic cells, thereby, preventing the production of inflammatory chemokines by CD40L-activated dendritic cells [97]. These studies demonstrate that GILZ is a glucocorticoid anti-inflammatory effect mediator and utilizes very similar mechanisms, to those GR uses [98].

Advertisement

9. GILZ mediates the anabolic effect of glucocorticoids

GILZ is a direct GR target gene with several GREs present in its promoter region [99]. In the absence of glucocorticoid stimulation GILZ is expressed at a very low basal level. However, in the presence of glucocorticoid, GILZ expression is rapidly induced (Figure 3] but GR is also activated, and the activated GR negatively impacts bone, both directly (i.e., inhibits osteocalcin gene transcription) and indirectly through other pathways as illustrated (Figure 4]. Because of that, it is impossible to determine the role of GILZ in osteoblast differentiation and bone formation without the influence of GR, which plays a negative role and may override GILZ actions. To further study this problem, a retrovirus-mediated GILZ overexpression system was established in bone marrow MSCs/osteoprogenitor cells. Studies carried out in this system showed that GILZ has potent pro-osteogenic activity as demonstrated by significantly increased alkaline phosphatase activity, enhanced mineralized bone nodule formation, and the expression of osteoblast-associated genes such as Runx2, type I collagen, alkaline phosphatase, and osteocalcin [100]. Furthermore, our recent studies have shown that overexpression of GILZ can antagonize the inhibitory effect of TNF-α on MSC osteogenic differentiation [101]. Possible mechanisms underlying this antagonism may include GILZ inhibition of TNF-α-induced ERK/MAP kinase activation, which has been shown to be responsible for TNF-α down-regulation of a key osteogenic factor Osx [102;103], and inhibition of TNF-α-induced expression of E3 ubiquitin ligase Smurf proteins, which have been shown to accelerate the degradation of Runx2 protein [104-106].

Figure 3.

GILZ enhances MSC osteogenic differentiation by shifting MSC lineage preference to osteogenic pathway.

Advertisement

10. GR mediates the catabolic effects of glucocorticoids

There are many glucocorticoid effectors involved in the regulation of bone development or metabolism through different pathways. However, it was only recently demonstrated that the GR was directly responsible for glucocorticoid-induced bone loss in vivo. Using a bone-specific GR knockout mouse model, Rauch et al showed that glucocorticoids are unable to induce bone loss or to inhibit bone formation in these mice because the GR-deficient osteoblasts become refractory to glucocorticoid-induced apoptosis, inhibition of proliferation, and differentiation [107]. Interestingly, data from this study also demonstrated that GR-deficiency results in a low bone mass phenotype, confirming the previous studies that the endogenous glucocorticoid signaling is critical for normal bone acquisition [54-56]. Other evidence supporting the role of GR in glucocorticoid-induced bone loss includes: 1] the glucocorticoid-activated GR binds directly to the negative glucocorticoid response elements (nGREs) in the promoter region of the osteocalcin (Ocn) gene, an osteoblast-specific gene that plays an important role in bone mineralization, and inhibit its transcription [36;108]; 2] GR transcriptionally activates the expression of MAP kinase phosphatase-1 (MKP-1] [109], which inactivates MAP kinase and thus inhibits osteogenic differentiation [64;110;111]; and 3] GR can physically interact with and inhibit the transcriptional functions of Smad3, an intracellular signaling mediator of transforming growth factor-beta (TGF-β) [112] (Figure 4]. Glucocorticoids have been known to antagonize TGF-β action in bone [113-115] and TGF-β stimulates osteoprogenitor cell proliferation [116-119] and attract osteoprogenitor cells to the remodeling sites during bone remodeling [120]. It is important to note that while the catabolic effects of glucocorticoids are often associated with long-term glucocorticoid excess [1-4;7], a short term exposure to glucocorticoid seems beneficial; for example, treatment of bone marrow stromal cells or osteoblasts with dexamethasone enhances, rather than inhibits, alkaline phosphatase (ALP) activity. The ALP is expressed at the early stage of osteoblast differentiation program and the increase of ALP expression or activity marks the entry of cells into the osteoblast lineage.

Figure 4.

GR inhibits MSC proliferation, ERK activation and Ocn expression. Ligand-bound GR physically interacts with: 1] TGF-β signaling mediator Smad3 and disrupts its transcriptional activity; 2] Activates mkp-1 transcription, by binding to GRE in the mkp-1 promoter region, resulting inhibition of ERK activation; and 3] Suppresses Ocn expression by binding to nGRE in the Ocn promoter region.

11. Glucocorticoid-Induced Osteoporosis (GIOP)

Although glucocorticoids are an essential hormone for survival and normal function, when present in excess (pharmacologic doses) lead to a number of serious side effects including bone loss and fractures. In fact, it is estimated that 30-50% of patients chronically exposed to high levels of glucocorticoids will develop a bone fracture [121]. Glucocorticoid excess can result from either endogenous (Cushing’s) or exogenous (iatrogenic) sources. Glucocorticoids are widely used for the treatment of a variety of inflammatory and autoimmune conditions. It is estimated that 0.5% of the population receives steroid therapy and exogenous steroids are thus the most common cause of secondary osteoporosis [122]. There has been a lot of discussion on the dose, duration and mode of administration of steroids and the impact on the development of osteoporosis. There has been a lot of debate on a “safe” dose for glucocorticoid replacement. Doses as low as 2.5 mg of prednisolone have been reported to result in osteoporosis [122]. Even patients on “physiologic” glucocorticoid replacement for Addison’s disease have been reported to have lower bone density than controls, although clearly many of these patients were overreplaced with steroid therapy [123]. Further, steroids even when given in an intermittent, rather than continuous fashion, or in an inhaled rather than oral fashion, are still associated with an increased risk of fracture. In treatment guidelines by the American College of Rheumatology in 2010, it was recommended that for patients with low fracture risk receiving more than 7.5 mg of prednisolone equivalents for more than 3 months receive some form of therapy for fracture prevention. In contrast for patients at high fracture risk it was recommended that they receive some form of therapy even at glucocorticoid doses lower than 5 mg and even for periods for less than one month [124].

12. Mechanism

Glucocorticoids have multiple effects on bone and bone cells. In addition, in cases where the glucocorticoids are given to treat systemic inflammatory conditions (e.g. rheumatoid arthritis), the underlying condition also contributes to bone loss. Glucocorticoids also inhibit endogenous production of sex steroids (testosterone and estrogen) in addition to production of adrenal androgens, all of which may have protective effects against bone loss [125]. Further, prolonged high dose glucocorticoid use results in both muscle weakness thus predisposing to an increased number of falls and muscle wasting. Bone-muscle interactions may also contribute to maintaining bone health. Glucocorticoids also decrease intestinal calcium absorption thus further predisposing to osteoporosis. Recently, effects of glucocorticoids on decreasing bone vasculature, has also been implicated as a potential mechanism for glucocorticoid effects on bone [126]. There also seems to be an age-dependence of glucocorticoid effects on bone. The likelihood of fractures with glucocorticoids appears to increase with increasing patient age. Glucocorticoid-induced bone loss appears to be biphasic with an initial rapid phase of bone loss of 5-15% /year followed by a more sustained bone loss rate of 2% [121].

Glucocorticoids affect all bone cells, they result in osteocytic and osteoblastic apoptosis and decreased function of both osteoclasts and osteoblasts. However, they decrease osteoclastic apoptosis. Thus, the net effect is reduced bone formation and increased bone breakdown. Trabecular bone seems to be particularly sensitive to the detrimental effects of steroids resulting in a higher incidence of vertebral and femoral neck fractures [121]. Vertebral compression fractures are commonly missed since only about 30% of them are symptomatic. A study by Angeli et al [127] which examined the prevalence of vertebral fractures in patients receiving glucocorticoids for a variety of autoimmune conditions determined that over 37% of patients had at least one asymptomatic vertebral compression fracture and more than 14% had two or more asymptomatic fractures.

Glucocorticoid effects on bone appear to be generally reversible and once therapy is stopped bone repair occurs over the year following drug cessation. Thus, if feasible, steroid cessation may be the therapy of choice for GIOP.

13. Diagnosis

Determining fracture risk for patients on steroids is difficult since even patients with normal bone densitometry on steroids have a higher fracture risk. Current use of steroids is one of the risk factors used in the calculation of the FRAX (Fracture Risk Assessment) score. However a recent joint position statement by the International Society for Clinical Densitometry and the International Osteoporosis Foundation concluded that when using the FRAX tool there probably was an underestimation of fracture risk with daily prednisone doses greater than 7.5 mg and an overestimation of fracture risk with daily prednisone doses of less than 2.5 mg. In addition, FRAX probably underestimated fracture risk when high dose inhaled steroids were used. Finally, it was concluded that for patients with adrenal insufficiency receiving appropriate replacement steroid doses this not be included in the FRAX calculation [128].

The American College of Rheumatology recommends that some form of therapy be considered for all patients receiving prolonged steroid therapy and that for those who have a bone densitometry test (Dual energy x-ray absorptiometry or DXA), a T-score of less than -1.0 be considered abnormal [125].

14. Therapy

Since glucocorticoids interfere with intestinal calcium absorption, all patients about to start glucocorticoid therapy should be placed on calcium and vitamin D replacement. Antiresorptive agents such as bisphosphonates (both oral and IV) have been used for the therapy of GIOP, are effective in decreasing the increased fracture risk associated with steroids and are approved for this indication. However, as discussed by Teitelbaum et al. [129], although initial use of steroids is associated with increased bone resorption (osteoclast mediated and related to decreased osteoclastic apoptosis and a situation in which antiresorptive use makes sense), more prolonged steroid use is associated with decreased bone formation and antiresorptive agents have the theoretical possibility of making things worse by further suppressing a low bone turnover state. Thus, use of an anabolic agent such as teriparatide (synthetic parathyroid hormone) for treatment of GIOP would appear more appropriate. In fact in a clinical trial, comparing alendronate vs. teriparatide for 18 months in 428 men/women with established osteoporosis and who had received at least 5mg of prednisone for at least 3 months, teriparatide was significantly more effective in both increasing bone mineral density at the spine [7.2 vs 3.4%) and in decreasing new vertebral fractures [0.6% vs 6.1%) [130]. Of note, this was a secondary instead of a primary osteoporosis prevention trial and there was a greater incidence of side effects associated with teriparatide use as compared to controls [131]. In addition, use of teriparatide by patients is currently limited to two years, thus alternative and better forms of therapy for GIOP need to be developed.

15. Conclusion

Although adverse side effects of glucocorticoids on bone have been long recognized, both from endogenous sources as described by Harvey Cushing in the 1930’s or from exogenous sources after development of glucocorticoids in the 1950’s the mechanisms involved in this process have only recently began to be understood. It is clear that although physiologic levels of glucocorticoids are important in normal bone development, pharmacologic doses result in a high level of fractures, particularly of vertebral bone. Thus, it would seem that glucocorticoids have both anabolic and catabolic actions on bone. Data from our labs and from others suggest that GILZ may be an important mediator of GR’s anabolic actions and thus may be an attractive therapeutic target for drug development.

Acknowledgement

This work was supported by grants from the National Institutes of Health (DK76045] and National Institute on Aging (P01AG036675].

References

  1. 1. HarrisE. DJr., Emkey,R.D., Nichols,J.E., and Newberg,A. 1983Low dose prednisone therapy in rheumatoid arthritis: a double blind study.J.Rheumatol. 10713721
  2. 2. ConnD. L2001Resolved: Low-dose prednisone is indicated as a standard treatment in patients with rheumatoid arthritis.Arthritis Rheum. 45462467
  3. 3. LocascioVBonucciEImbimboBBallantiPAdamiSMilaniSTartarottiDand DellaRocca,C. 1990Bone loss in response to long-term glucocorticoid therapy.Bone Miner. 83951
  4. 4. NishimuraJand IkuyamaS2000Glucocorticoid-induced osteoporosis: pathogenesis and management.J.Bone Miner.Metab 18350352
  5. 5. The Lung Health Study Research Group2000Effect of inhaled triamcinolone on the decline in pulmonary function in chronic obstructive pulmonary diseaseN.Engl.J.Med. 34319021909
  6. 6. WalshL. JWongC. AOborneJCooperSLewisS. APringleMHubbardRand TattersfieldA. E2001Adverse effects of oral corticosteroids in relation to dose in patients with lung diseaseThorax 56279284
  7. 7. RuegseggerPMediciT. Cand AnlikerM1983Corticosteroid-induced bone loss. A longitudinal study of alternate day therapy in patients with bronchial asthma using quantitative computed tomography.Eur.J.Clin.Pharmacol. 25615620
  8. 8. Bazzy-asaadA2001Safety of inhaled corticosteroids in children with asthma.Curr.Opin.Pediatr. 13523527
  9. 9. CorrenJNelsonHGreosL. SBenschGGoldsteinMWuJWangSand NewmanK2001Effective control of asthma with hydrofluoroalkane flunisolide delivered as an extrafine aerosol in asthma patients.Ann.Allergy Asthma Immunol. 87405411
  10. 10. FernandesA. LFaresinS. MAmorimM. MFritscherC. CPereiraC. Aand JardimJ. R2001Inhaled budesonide for adults with mild-to-moderate asthma: a randomized placebo-controlled, double-blind clinical trial.Sao Paulo Med.J. 119169174
  11. 11. AdinoffA. Dand HollisterJ. R1983Steroid-induced fractures and bone loss in patients with asthma.N.Engl.J.Med. 309265268
  12. 12. ParkS. JNguyenD. QSavikKHertzM. Iand BolmanR. MIII. 2001Pre-transplant corticosteroid use and outcome in lung transplantation.J.Heart Lung Transplant. 20304309
  13. 13. DequekerJand WesthovensR1995Low dose corticosteroid associated osteoporosis in rheumatoid arthritis and its prophylaxis and treatment: bones of contention.J.Rheumatol. 2210131019
  14. 14. AdachiJ. DOlszynskiW. PHanleyD. AHodsmanA. BKendlerD. LSiminoskiK. GBrownJCowdenE. AGoltzmanDIoannidisGet al2000Management of corticosteroid-induced osteoporosis. Semin.Arthritis Rheum. 29228251
  15. 15. SaagK. GKoehnkeRCaldwellJ. RBrasingtonRBurmeisterL. FZimmermanBKohlerJ. Aand FurstD. E1994Low dose long-term corticosteroid therapy in rheumatoid arthritis: an analysis of serious adverse events.Am.J.Med. 96115123
  16. 16. BraunJand SieperJ2001Glucocorticoid-induced osteoporosis]. Orthopade 30444450
  17. 17. ClowesJ. APeelNand EastellR2001Glucocorticoid-induced osteoporosis.Curr.Opin.Rheumatol. 13326332
  18. 18. LukertB. Pand RaiszL. G1990Glucocorticoid-induced osteoporosis: pathogenesis and management.Ann.Intern.Med. 112352364
  19. 19. KleinG2004Glucocorticoid-induced bone loss in childrenClinical Reviews in Bone and Mineral Metabolism 23752
  20. 20. Van StaaT. PCooperCLeufkensHand BishopN2003Children and the Risk of Fractures Caused by Oral CorticosteroidsJournal of Bone and Mineral Research 18913918
  21. 21. BootA. MDe JongsteJ. CVerberneA. A. P. HPolsH. A. Pand de Muinck Keizer-Schrama,S. 1997Bone mineral density and bone metabolism of prepubertal children with asthma after long-term treatment with inhaled corticosteroids.Pediatr.Pulmonol. 24379384
  22. 22. CovarR. ALeungD. YMccormickDSteelmanJZeitlerPand SpahnJ. D2000Risk factors associated with glucocorticoid-induced adverse effects in children with severe asthmaJ Allergy Clin Immunol 106651659
  23. 23. BurnhamJ. Mand LeonardM. B2004Bone disease in pediatric rheumatologic disordersCurr.Rheumatol.Rep. 67078
  24. 24. ViswanathanAand SylvesterF2008Chronic pediatric inflammatory diseases: Effects on boneReviews in Endocrine & Metabolic Disorders9107122
  25. 25. BurnhamJ. MShultsJSemeaoEFosterBZemelB. SStallingsV. Aand LeonardM. B2004Whole Body BMC in Pediatric Crohn Disease: Independent Effects of Altered Growth, Maturation, and Body Composition.Journal of Bone and Mineral Research 1919611968
  26. 26. LillebyVLienGFrey Fr_Ėslie,K., Haugen,M., Flat_Ė,B., and F_Ėrre,ś. 2005Frequency of osteopenia in children and young adults with childhood-onset systemic lupus erythematosus. Arthritis & Rheumatism 5220512059
  27. 27. Compeyrot-lacassagneSTyrrellP. NAtenafuEDoriaA. SStephensDGildayDand SilvermanE. D2007Prevalence and etiology of low bone mineral density in juvenile systemic lupus erythematosus.Arthritis & Rheumatism 5619661973
  28. 28. WaltherFFuschCRadkeMBeckertSand FindeisenA2006Osteoporosis in pediatric patients suffering from chronic inflammatory bowel disease with and without steroid treatment.J Pediatr.Gastroenterol.Nutr. 434251
  29. 29. BootA. MBouquetJKrenningE. Pand Muinck Keizer-Schrama,S.M.P.F. 1998Bone mineral density and nutritional status in children with chronic inflammatory bowel diseaseGut 42188194
  30. 30. MazziottiGAngeliABilezikianJ. PCanalisEand GiustinaA2006Glucocorticoid-induced osteoporosis: an updateTrends in Endocrinology & Metabolism 17144149
  31. 31. WebsterJ. Cand CidlowskiJ. A1999Mechanisms of Glucocorticoid-receptor-mediated Repression of Gene ExpressionTrends in Endocrinology and Metabolism 10396402
  32. 32. KumarRand ThompsonE. B2005Gene regulation by the glucocorticoid receptor: Structure:function relationshipThe Journal of Steroid Biochemistry and Molecular Biology94383394
  33. 33. KallioP. JPalvimoJand JanneO. A1994Nuclear hormone receptors]. Duodecim 110383394
  34. 34. GiguereVHollenbergS. MRosenfeldM. Gand EvansR. M1986Functional domains of the human glucocorticoid receptor.Cell46645652
  35. 35. ParkerM. G1990Structure and function of nuclear hormone receptors. Semin.Cancer Biol 18187
  36. 36. MorrisonNand EismanJ1993Role of the negative glucocorticoid regulatory element in glucocorticoid repression of the human osteocalcin promoter.J Bone Miner.Res. 8969975
  37. 37. De BosscherKVanden Berghe,W., and Haegeman,G. 2003The Interplay between the Glucocorticoid Receptor and Nuclear Factor-{kappa}B or Activator Protein-1: Molecular Mechanisms for Gene Repression. Endocr Rev 24488522
  38. 38. De BosscherKVanden Berghe,W., and Haegeman,G. 2000Mechanisms of anti-inflammatory action and of immunosuppression by glucocorticoids: negative interference of activated glucocorticoid receptor with transcription factors.J Neuroimmunol. 1091622
  39. 39. ZitnikR. JWhitingN. Land EliasJ. A1994Glucocorticoid inhibition of interleukin-1-induced interleukin-6 production by human lung fibroblasts: evidence for transcriptional and post-transcriptional regulatory mechanisms.Am J Respir.Cell Mol Biol 10643650
  40. 40. Yang-yenH. FChambardJ. CSunY. LSmealTSchmidtT. JDrouinJand KarinM1990Transcriptional interference between c-Jun and the glucocorticoid receptor: mutual inhibition of DNA binding due to direct protein-protein interaction.Cell6212051215
  41. 41. JonatCRahmsdorfH. JParkK. KCatoA. CGebelSPontaHand HerrlichP1990Antitumor promotion and antiinflammation: down-modulation of AP-1 (Fos/Jun) activity by glucocorticoid hormone.Cell6211891204
  42. 42. SchuleRRangarajanPKliewerSRansoneL. JBoladoJYangNVermaI. Mand EvansR. M1990Functional antagonism between oncoprotein c-Jun and the glucocorticoid receptor.Cell6212171226
  43. 43. RayAand PrefontaineK. E1994Physical association and functional antagonism between the 65subunit of transcription factor NF-kappa B and the glucocorticoid receptor.Proc.Natl.Acad.Sci.U.S.A 91:752-756.
  44. 44. ScheinmanR. IGualbertoAJewellC. MCidlowskiJ. Aand BaldwinA. SJr. 1995Characterization of mechanisms involved in transrepression of NF-kappa B by activated glucocorticoid receptors.Mol.Cell.Biol. 15943953
  45. 45. AuphanNDiDonato,J.A., Rosette,C., Helmberg,A., and Karin,M. 1995Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis.Science 270286290
  46. 46. ScheinmanR. ICogswellP. CLofquistA. Kand BaldwinA. SJr. 1995Role of transcriptional activation of I kappa B alpha in mediation of immunosuppression by glucocorticoids.Science 270283286
  47. 47. CrinelliRAntonelliABianchiMGentiliniLScaramucciSand MagnaniM2000Selective inhibition of NF-kB activation and TNF-alpha production in macrophages by red blood cell-mediated delivery of dexamethasone.Blood CellsMol Dis 26211222
  48. 48. ThieleKBierhausAAutschbachFHofmannMStremmelWThieleHZieglerRand NawrothP. P1999Cell specific effects of glucocorticoid treatment on the NF-kappaBp65/IkappaBalpha system in patients with Crohn’s disease.Gut45693704
  49. 49. De BosscherKScmitzM. LVanden Berghe, W., Plaisance, S.p., Fiers, W., and Haegeman, G. 1997Glucocorticoid-mediated repression of nuclear factor-dependent transcription involves direct interference with transactivation. PNAS 941350413509
  50. 50. BrostjanCAnratherJCsizmadiaVStrokaDSoaresMBachF. Hand WinklerH1996Glucocorticoid-mediated Repression of NF__B Activity in Endothelial Cells Does Not Involve Induction of I__B__ Synthesis. J.Biol.Chem. 2711961219616
  51. 51. Heck,S., Bender,K., Kullmann,M., Gottlicher,M., Herrlich,P., and Cato,A.C. 1997. I kappaB alpha-independent downregulation of NF-kappaB activity by glucocorticoid receptor. EMBO J 16:4698-4707.
  52. 52. CanalisEMazziottiGGiustinaAand BilezikianJ. P2007Glucocorticoid-induced osteoporosis: pathophysiology and therapyOsteoporos.Int 1813191328
  53. 53. LukertB. Pand RaiszL. G1994Glucocorticoid-induced osteoporosis. Rheum.Dis.Clin.North Am. 20629650
  54. 54. SherL. BWoitgeH. WAdamsD. JGronowiczG. AKrozowskiZHarrisonJ. Rand KreamB. E2004Transgenic Expression of 11__-Hydroxysteroid Dehydrogenase Type 2 in Osteoblasts Reveals an Anabolic Role for Endogenous Glucocorticoids in Bone. Endocrinology145922929
  55. 55. SherL. BHarrisonJ. RAdamsD. Jand KreamB. E2006Impaired cortical bone acquisition and osteoblast differentiation in mice with osteoblast-targeted disruption of glucocorticoid signalingCalcif.Tissue Int 79118125
  56. 56. YangMTrettelL. BAdamsD. JHarrisonJ. RCanalisEand KreamB. E2010Col3.6-HSD2 transgenic mice: A glucocorticoid loss-of-function model spanning early and late osteoblast differentiationBone47573582
  57. 57. BruderS. PJaiswalNand HaynesworthS. E1997Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation.J.Cell Biochem. 64278294
  58. 58. EnglerA. JSenSSweeneyH. Land DischerD. E2006Matrix Elasticity Directs Stem Cell Lineage SpecificationCell126677689
  59. 59. PittengerM. FMackayA. MBeckS. CJaiswalR. KDouglasRMoscaJ. DMoormanM. ASimonettiD. WCraigSand MarshakD. R1999Multilineage Potential of Adult Human Mesenchymal Stem Cells.Science 284143147
  60. 60. ProckopD. J1997Marrow stromal cells as stem cells for nonhematopoietic tissuesScience2767174
  61. 61. AhdjoudjSLasmolesFOyajobiB. OLomriADelannoyPand MarieP. J2001Reciprocal control of osteoblast/chondroblast and osteoblast/adipocyte differentiation of multipotential clonal human marrow stromal F/STRO-1(+) cells. J Cell Biochem. 812338
  62. 62. BeresfordJ. NBennettJ. HDevlinCLeboyP. Sand OwenM. E1992Evidence for an inverse relationship between the differentiation of adipocytic and osteogenic cells in rat marrow stromal cell cultures.J.Cell Sci. 102 ( Pt 2]:341351
  63. 63. GoriFThomasTHicokK. CSpelsbergT. Cand RiggsB. L1999Differentiation of human marrow stromal precursor cells: bone morphogenetic protein-2 increases OSF2/CBFA1, enhances osteoblast commitment, and inhibits late adipocyte maturation.J.Bone Miner.Res. 1415221535
  64. 64. JaiswalR. KJaiswalNBruderS. PMbalavieleGMarshakD. Rand PittengerM. F2000Adult human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by mitogen-activated protein kinase.J.Biol.Chem. 27596459652
  65. 65. BarakYNelsonM. COngE. SJonesY. ZRuiz-lozanoPChienK. RKoderAand EvansR. M1999PPAR gamma is required for placental, cardiac, and adipose tissue development.Mol.Cell 4585595
  66. 66. RosenE. DSarrafPTroyA. EBradwinGMooreKMilstoneD. SSpiegelmanB. Mand MortensenR. M1999PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro.Mol.Cell 4611617
  67. 67. ShiX. MBlairH. CYangXMcdonaldJ. Mand CaoX2000Tandem repeat of C/EBP binding sites mediates PPARgamma2 gene transcription in glucocorticoid-induced adipocyte differentiation.J.Cell Biochem. 76518527
  68. 68. WuZBucherN. Land FarmerS. R1996Induction of peroxisome proliferator-activated receptor gamma during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPbeta, C/EBPdelta, and glucocorticoids.Mol.Cell.Biol. 1641284136
  69. 69. TangQ. QZhangJ. Wand Daniel Lane,M. 2004Sequential gene promoter interactions by C/EBP[beta], C/EBP[alpha], and PPAR[gamma] during adipogenesis. Biochemical and Biophysical Research Communications318213218
  70. 70. ClarkeS. LRobinsonC. Eand GimbleJ. M1997CAAT/Enhancer Binding Proteins Directly Modulate Transcription from the Peroxisome Proliferator- Activated Receptor [gamma]2 Promoter. Biochemical and Biophysical Research Communications24099103
  71. 71. WeinsteinR. SJilkaR. LParfittA. Mand ManolagasS. C1998Inhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms of their deleterious effects on bone.J Clin.Invest 102274282
  72. 72. YaoWChengZBusseCPhamANakamuraM. Cand LaneN. E2008Glucocorticoid excess in mice results in early activation of osteoclastogenesis and adipogenesis and prolonged suppression of osteogenesis: A longitudinal study of gene expression in bone tissue from glucocorticoid-treated mice.Arthritis & Rheumatism 5816741686
  73. 73. AkuneTOhbaSKamekuraSYamaguchiMChungU. IKubotaNTerauchiYHaradaYAzumaYNakamuraKet al2004PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors.J.Clin.Invest 113846855
  74. 74. CanalisE2005Mechanisms of glucocorticoid action in bone.Curr.Osteoporos.Rep. 398102
  75. 75. WeinsteinR. S2001Glucocorticoid-induced osteoporosis.Rev.Endocr.Metab Disord. 26573
  76. 76. OBrienC. AJiaDPlotkinL. IBellidoTPowersC. CStewartS. AManolagasS. Cand WeinsteinR.S. 2004Glucocorticoids Act Directly on Osteoblasts and Osteocytes to Induce Their Apoptosis and Reduce Bone Formation and Strength.Endocrinology14518351841
  77. 77. RoblingA. GNiziolekP. JBaldridgeL. ACondonK. WAllenM. RAlamIMantilaS. MGluhak-heinrichJBellidoT. MHarrisS. Eet al2008Mechanical Stimulation of Bone in Vivo Reduces Osteocyte Expression of Sost/SclerostinJ.Biol.Chem. 28358665875
  78. 78. Gluhak-heinrichJYeLBonewaldL. FFengJ. QMacDougall,M., Harris,S.E., and Pavlin,D. 2003Mechanical Loading Stimulates Dentin Matrix Protein 1 (DMP1] Expression in Osteocytes In Vivo.Journal of Bone and Mineral Research 18807817
  79. 79. HirayamaTSabokbarAand AthanasouN. A2002Effect of corticosteroids on human osteoclast formation and activity.J Endocrinol 175155163
  80. 80. SivagurunathanSMuirM. MBrennanT. CSealeJ. Pand MasonR. S2005Influence of Glucocorticoids on Human Osteoclast Generation and Activity.Journal of Bone and Mineral Research 20390398
  81. 81. JiaDOBrienC. AStewartS. AManolagasS. Cand WeinsteinR.S. 2006Glucocorticoids Act Directly on Osteoclasts to Increase Their Life Span and Reduce Bone Density.Endocrinology14755925599
  82. 82. KimH. JZhaoHKitauraHBhattacharyyaSBrewerJ. AMugliaL. JRossF. Pand TeitelbaumS. L2006Glucocorticoids suppress bone formation via the osteoclastJ Clin Invest 11621522160
  83. 83. SEeKand DelaissJ.M. 2010Glucocorticoids maintain human osteoclasts in the active mode of their resorption cycle. Journal of Bone and Mineral Research 2521842192
  84. 84. DAdamioFZolloOMoracaRAyroldiEBruscoliSBartoliACannarileLMiglioratiGand RiccardiC. 1997A new dexamethasone-induced gene of the leucine zipper family protects T lymphocytes from TCR/CD3-activated cell death.Immunity7803812
  85. 85. CannarileLZolloODAdamioFAyroldiEMarchettiCTabilioABruscoliSand RiccardiC2001Cloningchromosomal assignment and tissue distribution of human GILZ, a glucocorticoid hormone-induced gene. Cell Death.Differ. 8201203
  86. 86. AyroldiEZolloOMacchiaruloADi Marco,B., Marchetti,C., and Riccardi,C. 2002Glucocorticoid-induced leucine zipper inhibits the Raf-extracellular signal-regulated kinase pathway by binding to Raf-1.Mol.Cell Biol. 2279297941
  87. 87. ShibanumaMKurokiTand NoseK1992Isolation of a gene encoding a putative leucine zipper structure that is induced by transforming growth factor beta 1 and other growth factors.J.Biol.Chem. 2671021910224
  88. 88. NewtonRHartL. AStevensD. ABergmannMDonnellyL. EAdcockI. Mand BarnesP. J1998Effect of dexamethasone on interleukin-1beta-(IL-1beta)-induced nuclear factor-kappaB (NF-kappaB) and kappaB-dependent transcription in epithelial cells. Eur J Biochem 2548189
  89. 89. EddlestonJHerschbachJWagelie-steffenA. LChristiansenS. Cand ZurawB. L2007The anti-inflammatory effect of glucocorticoids is mediated by glucocorticoid-induced leucine zipper in epithelial cellsJ Allergy Clin Immunol 119115122
  90. 90. DelfinoD. VAgostiniMSpinicelliSVitoPand RiccardiC2004Decrease of Bcl-xL and augmentation of thymocyte apoptosis in GILZ overexpressing transgenic mice.Blood
  91. 91. AyroldiEMiglioratiGBruscoliSMarchettiCZolloOCannarileLDAdamioFand RiccardiC. 2001Modulation of T-cell activation by the glucocorticoid-induced leucine zipper factor via inhibition of nuclear factor kappaB.Blood98743753
  92. 92. MittelstadtP. Rand AshwellJ. D2001Inhibition of AP-1 by the glucocorticoid-inducible protein GILZ.J.Biol.Chem. 2762960329610
  93. 93. AyroldiEZolloOBastianelliAMarchettiCAgostiniMDi Virgilio,R., and Riccardi,C. 2007GILZ mediates the antiproliferative activity of glucocorticoids by negative regulation of Ras signalingJ Clin Invest 11716051615
  94. 94. BerrebiDBruscoliSCohenNFoussatAMiglioratiGBouchet-delbosLMaillotM. CPortierACoudercJGalanaudPet al2003Synthesis of glucocorticoid-induced leucine zipper (GILZ) by macrophages: an anti-inflammatory and immunosuppressive mechanism shared by glucocorticoids and IL-10.Blood101729738
  95. 95. YangNZhangWand ShiX. M2007Glucocorticoid-induced leucine zipper (GILZ) mediates glucocorticoid action and inhibits inflammatory cytokine-induced COX-2 expressionJ Cell Biochem 10317601771
  96. 96. ArthaningtyasEKokC. CMordvinovV. Aand SandersonC. J2005The conserved lymphokine element 0 is a powerful activator and target for corticosteroid inhibition in human interleukin-5 transcriptionGrowth Factors23211221
  97. 97. CohenNMoulyEHamdiHMaillotM. CPallardyMGodotVCapelFBalianANaveauSGalanaudPet al2005GILZ expression in human dendritic cells redirects their maturation and prevents antigen-specific T lymphocyte response.Blood20052007
  98. 98. AyroldiEand RiccardiC2009Glucocorticoid-induced leucine zipper (GILZ): a new important mediator of glucocorticoid actionFASEB J. 23110
  99. 99. Asselin-Labat,M.L., Biola-Vidamment,A., Kerbrat,S., Lombes,M., Bertoglio,J., and Pallardy,M. 2005. FoxO3 Mediates Antagonistic Effects of Glucocorticoids and Interleukin-2 on Glucocorticoid-Induced Leucine Zipper Expression. Mol Endocrinol 19:1752-1764.
  100. 100. ZhangWYangNand ShiX. M2008Regulation of Mesenchymal Stem Cell Osteogenic Differentiation by Glucocorticoid-induced Leucine Zipper (GILZ)J.Biol.Chem. 28347234729
  101. 101. HeLYangNIsalesC. Mand ShiX. M2012Glucocorticoid-Induced Leucine Zipper (GILZ) Antagonizes TNF-__ Inhibition of Mesenchymal Stem Cell Osteogenic Differentiation. PLoS ONE 7:e31717.
  102. 102. NakashimaKZhouXKunkelGZhangZ. PDengJ. MBehringerR. Rand De CrombruggheB2002The novel zinc finger-containing transcription factor Osterix is required for osteoblast differentiation and bone formation.Cell1081729
  103. 103. LuXGilbertLHeXRubinJand NanesM. S2006Transcriptional Regulation of the Osterix (Osx, Sp7] Promoter by Tumor Necrosis Factor Identifies Disparate Effects of Mitogen-activated Protein Kinase and NF+¦B Pathways. J.Biol.Chem. 28162976306
  104. 104. GilbertLHeXFarmerPRubinJDrissiHVan WijnenA. JLianJ. BSteinG. Sand NanesM. S2002Expression of the Osteoblast Differentiation Factor RUNX2 (Cbfa1/AML3/Pebp2alpha A) Is Inhibited by Tumor Necrosis Factor-alpha.J.Biol.Chem. 27726952701
  105. 105. KanekiHGuoRChenDYaoZSchwarzE. MZhangY. EBoyceB. Fand XingL2006Tumor Necrosis Factor Promotes Runx2 Degradation through Up-regulation of Smurf1 and Smurf2 in Osteoblasts.J.Biol.Chem. 28143264333
  106. 106. ZhaoMQiaoMOyajobiB. OMundyG. Rand ChenD2003E3 ubiquitin ligase Smurf1 mediates core-binding factor alpha1/Runx2 degradation and plays a specific role in osteoblast differentiation.J.Biol.Chem. 2782793927944
  107. 107. RauchASeitzSBaschantUSchillingA. FIllingAStrideBKirilovMMandicVTakaczASchmidt-ullrichRet al2010Glucocorticoids Suppress Bone Formation by Attenuating Osteoblast Differentiation via the Monomeric Glucocorticoid ReceptorCell Metabolism11517531
  108. 108. YaoWChengZBusseCPhamANakamuraM. Cand LaneN. E2008Glucocorticoid excess in mice results in early activation of osteoclastogenesis and adipogenesis and prolonged suppression of osteogenesis: A longitudinal study of gene expression in bone tissue from glucocorticoid-treated mice.Arthritis & Rheumatism 5816741686
  109. 109. KasselOSanconoAKratzschmarJKreftBStassenMand CatoA. C. B2001Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1.EMBO J 2071087116
  110. 110. SuzukiAGuicheuxJPalmerGMiuraYOisoYBonjourJ. Pand CaverzasioJ2002Evidence for a role of 38MAP kinase in expression of alkaline phosphatase during osteoblastic cell differentiation.Bone
  111. 111. ParkO. JKimH. JWooK. MBaekJ. Hand RyooH. M2010FGF2-activated ERK Mitogen-activated Protein Kinase Enhances Runx2 Acetylation and StabilizationJ.Biol.Chem. 28535683574
  112. 112. SongC. ZTianXand GelehrterT. D1999Glucocorticoid receptor inhibits transforming growth factor-beta signaling by directly targeting the transcriptional activation function of Smad3.Proc Natl Acad Sci U.S.A 961177611781
  113. 113. IuM. FKajiHSowaHNaitoJSugimotoTand ChiharaK2005Dexamethasone suppresses Smad3 pathway in osteoblastic cells.J Endocrinol 185131138
  114. 114. PeriyasamySand SanchezE. R2002Antagonism of glucocorticoid receptor transactivity and cell growth inhibition by transforming growth factor-[beta] through AP-1-mediated transcriptional repression. The International Journal of Biochemistry & Cell Biology3415711585
  115. 115. SongC. ZTianXand GelehrterT. D1999Glucocorticoid receptor inhibits transforming growth factor-beta signaling by directly targeting the transcriptional activation function of Smad3.Proc Natl Acad Sci U.S.A 961177611781
  116. 116. LocklinR. MOreffoR. O. Cand TriffittJ. T1999Effects of TGF[beta] and BFGF on the differentiation of human bone marrow stromal fibroblasts. Cell Biology International23185194
  117. 117. AllistonTChoyLDucyPKarsentyGand DerynckR2001TGF-[beta]-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. EMBO J 2022542272
  118. 118. KassemKveiborg, and Eriksen. 2000Production and action of transforming growth factor-__ in human osteoblast cultures: dependence on cell differentiation and modulation by calcitriol.European Journal of Clinical Investigation30429437
  119. 119. EdwardsJ. RNymanJ. SLwinS. TMooreM. MEsparzaJOQuinnE. CHartA. JBiswasSPatilC. ALonningS. et al2010Inhibition of TGF-beta signaling by 1D11 antibody treatment increases bone mass and quality in vivo. jbmrn-a.
  120. 120. TangYWuXLeiWPangLWanCShiZZhaoLNagyT. RPengXHuJet al2009TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation.Nat.Med. 15757765
  121. 121. BouvardBAudranMLegrandEand ChappardD2009Ultrastructural characteristics of glucocorticoid-induced osteoporosisOsteoporosis International2010891092
  122. 122. BerrisK. KReppA. Land KleerekoperM2007Glucocorticoid-induced osteoporosis.Current Opinion in EndocrinologyDiabetes and Obesity 14.
  123. 123. LEvŹsKGjesdalC. GChristensenMWolffA. BAlm_źs,B.Ė., Svartberg,J., Fougner,K.J., Syversen,U., Bollerslev,J., Falch,J.A. et al. 2009Glucocorticoid replacement therapy and pharmacogenetics in Addison’s disease: effects on bone. Eur J Endocrinol 1609931002
  124. 124. GrossmanJ. MGordonRRanganathV. KDealCCaplanLChenWCurtisJ. RFurstD. EMcmahonMPatkarN. Met al2010American College of Rheumatology 2010 recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis.Arthritis Care Res 6215151526
  125. 125. Van StaaT2006The Pathogenesis, Epidemiology and Management of Glucocorticoid-Induced Osteoporosis. Calcified Tissue International 79129137
  126. 126. WeinsteinR. S2010Glucocorticoids, osteocytes, and skeletal fragility: The role of bone vascularity. Bone 46564570
  127. 127. AngeliAGuglielmiGDovioACapelliGDe FeoDGianniniSGiorginoRMoroLand GiustinaA2006High prevalence of asymptomatic vertebral fractures in post-menopausal women receiving chronic glucocorticoid therapy: A cross-sectional outpatient studyBone39253259
  128. 128. LeibE. SSaagK. GAdachiJ. DGeusensP. PBinkleyNMccloskeyE. Vand HansD. B2011Official Positions for FRAX_« Clinical Regarding Glucocorticoids: The Impact of the Use of Glucocorticoids on the Estimate by FRAX_« of the 10 Year Risk of Fracture: From Joint Official Positions Development Conference of the International Society for Clinical Densitometry and International Osteoporosis Foundation on FRAX_«.Journal of Clinical Densitometry14212219
  129. 129. TeitelbaumS. LSetonM. Pand SaagK. G2011Should bisphosphonates be used for long-term treatment of glucocorticoid-induced osteoporosis? Arthritis & Rheumatism 63325328
  130. 130. SaagK. GShaneEBoonenSMar_Łn,F., Donley,D.W., Taylor,K.A., Dalsky,G.P., and Marcus,R. 2007Teriparatide or Alendronate in Glucocorticoid-Induced Osteoporosis.New England Journal of Medicine 35720282039
  131. 131. SambrookP. N2007Anabolic Therapy in Glucocorticoid-Induced Osteoporosis.New England Journal of Medicine 35720842086

Written By

Xing-Ming Shi, Norman Chutkan, Mark W. Hamrick and Carlos M. Isales

Submitted: 23 May 2012 Published: 28 November 2012