Skip to main content
Molecular Endocrinology logoLink to Molecular Endocrinology
. 2009 Oct;23(10):1525–1531. doi: 10.1210/me.2009-0069

Minireview: Live and Let Die: Molecular Effects of Glucocorticoids on Bone Cells

Lorenz C Hofbauer 1, Martina Rauner 1
PMCID: PMC5419139  PMID: 19477950

Abstract

Glucocorticoids (GCs) are efficient drugs that are used to treat various immune-mediated diseases, but their long-term administration is associated with multiple metabolic side effects, including osteoporosis. Molecular analyses of the mechanisms exerted by the GC receptor have resulted in the development of GC receptor agonists that selectively repress or activate GC target genes. This review summarizes the cellular and molecular effects of GCs on bone cells and highlights the critical signaling pathways that may evolve into future therapeutic strategies.


While traditional glucocorticoids have the drawback of impairing bone cell function through several distinct molecular mechanisms, newer selective receptor agonists may have bone-sparing effects.


Glucocorticoids (GCs) are used as potent immunomodulatory drugs for a broad range of immune-mediated diseases such as rheumatoid arthritis, asthma, and multiple sclerosis. The prolonged application of high doses of GCs, however, is limited by deleterious adverse effects on multiple organ systems, including the skeleton (1). Severe bone loss occurs within the first months of systemic GC use, and long-term GC treatment results in a slower but persistent decrease in bone mass and quality, leading to an increased risk of fractures (2). Up to 50% of the patients develop vertebral fractures under prolonged use of systemic GCs (3, 4). Although studies have shown that bone mineral density increases again after cessation of GC therapy, persistent inflammatory activity of the underlying disease frequently does not allow complete discontinuation of GCs (5).

Cells of the skeletal system (osteoclasts, osteoblasts, and osteocytes) express GC receptors (GRs) and GC-modifying enzymes that determine the susceptibility to GCs. Previous research on GC actions through the classical GR has highlighted the existence of various distinct pathways that either activate or repress target genes. The fact that these pathways may be selectively activated has resulted in the development of selective GR agonists (SEGRAs) that target the GR and are able to dissociate the immunosuppressive actions from some of the adverse effects. This review focuses on the current cellular and molecular understanding of GC effects on bone cells, the underlying pathways, and the bone-protective potential of SEGRAs.

Physiological bone remodeling

Bone mass and bone quality are maintained by continuous remodeling, a physiological process that adapts bone to changing functional, mechanical, and metabolic demands. The remodeling process is balanced between cells from the osteoblastic and osteoclastic lineages. Osteoblasts derive from mesenchymal stem cells and primarily secrete a collagenous bone matrix and orchestrate its subsequent mineralization. Osteocytes are terminally differentiated osteoblasts with characteristic dendrites. Using gap junctions that are located on their dendrites, osteocytes form a network throughout the bone tissue that allows them to communicate, to sense bone damage, and to direct its repair (6). Osteoclasts, on the other hand, are of hematopoietic origin and resorb bone by creating an acidic environment and producing collagen-degrading enzymes. Whereas receptor activator of nuclear factor-κB ligand (RANKL) is an essential cytokine for osteoclastogenesis, osteoprotegerin (OPG) acts as a decoy receptor for RANKL and suppresses osteoclastogenesis (7). Intriguingly, both proteins are produced by osteoblasts, thereby underlining the intimate communication between osteoblasts and osteoclasts in the cyclic process of bone remodeling (8).

The GRs and their actions

In principle, GCs exert their biological actions through genomic mechanisms using the GR. Although nongenomic mechanisms, i.e. interactions with cellular membranes and signal transduction via membrane-bound GRs have been reported, their relevance on bone cells remains currently elusive (9). Thus, this review will only focus on genomic mechanisms.

Alternative splicing of exon 9 of the human GR results in the production of two isoforms, GRα and GRβ. Bone cells express both GRs, but GRα is more abundantly expressed than GRβ (10).

GRα

GC actions mediated through the classical receptor GRα are well defined, and studies over the last decade have yielded important mechanistic insights. The unligated receptor is located in the cytoplasm and is part of a multiprotein complex that contains several heat-shock proteins, MAPKs, and chaperones. Ligation of GRα leads to its dissociation from the multiprotein complex. After translocation into the nucleus, GRα modifies gene expression through multiple pathways. To classify the various entities of GRα-mediated mechanisms, the terms “transactivation” and “transrepression” were coined. Accordingly, genes are activated by direct binding of GRα homodimers to GC response elements (e.g. phosphoenolpyruvate carboxykinase) or other DNA-bound transcription factors (e.g. signal transducer and activator of transcription 5). Transrepression, on the other hand, occurs upon binding to negative GC response elements (e.g. osteocalcin) or the interaction of GRα monomers with transcription factors (e.g. Smad3 in the case of type I collagen), coactivators, or corepressors (1, 11). Because many proteins that regulate metabolic functions are activated by GCs, transactivation has been suggested to account for most of the adverse effects. By contrast, transrepression inhibits many inflammatory mediators (activator protein 1, nuclear factor-κB) and is thought to mediate most of the desired antiinflammatory GC effects. As a caveat, however, this model does not hold true for all GC-regulated genes, because some genes that contribute to the antiinflammatory effects of GCs e.g. type II IL-1 receptor or lipocortin, are transactivated, whereas other genes that contribute to metabolic processes, e.g. osteocalcin are transrepressed.

GRβ

GRβ differs from the classical receptor GRα in that it has a distinct sequence of helix 11 and lacks helix 12 of the ligand-binding domain. Thus, earlier studies reported that GRβ does not bind agonists and only affects gene transcription through blocking the actions of GRα. However, this concept has been challenged by a recent study showing that, after binding of certain ligands such as the GR antagonist RU-486, GRβ is able to translocate into the nucleus independently of GRα and modify the gene expression pattern of target cells (12). Although little is known about the function and regulation of the GRβ in bone or other cells, clinical and in vitro studies propose a functional role of GRβ as a determinant of GC sensitivity because its expression correlates with GC resistance in patients with inflammatory conditions and cancer (13, 14). Thus, GRβ may play a role in fine tuning intraindividual GC susceptibility, but further studies are required to assess this in detail.

Pathogenesis of GC-inducedosteoporosis (GIO)

The pathogenesis of GIO is complex and involves many organ systems. GCs impair bone metabolism through extraskeletal effects including decreased calcium resorption in the gut, increased calcium loss through the kidneys, a decreased production and/or action of sex steroids and GH, decreased muscle strength, and impaired musculoskeletal interactions, all of which contribute to loss of bone mass, decreased bone quality, and a higher propensity to falls (15).

In addition, GCs have profound (physiological and pathological) direct effects on all cell types found in bone tissue. GCs suppress bone formation and stimulate bone resorption. Histomorphometric studies of patients and experimental animal models revealed a reduced bone formation rate and trabecular wall thickness due to reduced numbers of osteoblasts as well as enhanced apoptosis of mature osteoblasts and osteocytes (16, 17). Although the number of osteoclasts and parameters of bone erosion are within the normal range or slightly above, GCs have been shown to extend the life span of osteoclasts, providing a possible explanation for the sustained bone loss during prolonged GC treatment.

GC effects on the osteoblastic lineage

In contrast to physiological amounts of endogenous GCs that are necessary for efficient osteoblast differentiation (18, 19), GC excess shifts the osteoblast-adipocyte balance toward enhanced adipogenesis (Fig. 1, orange box), inhibits osteoblast differentiation and function (blue box), and increases both osteoblast and osteocyte apoptosis (blue and green box).

Fig. 1.

Fig. 1.

Cellular effects and molecular pathways employed by GCs in the homeostasis of bone. GCs, as illustrated by cortisol, enhance the expression of adipogenic and osteogenic transcription factors in mesenchymal stem cells. Long-term exposure may increase adipogenesis (orange). The GC-mediated arrest of various signaling pathways, including IGF-1, TGF-β/bone morphogenetic protein, and Wnt suppresses the osteoblastic transcription factors Runx2 and β-catenin and attenuates osteoblast differentiation. Arrest of osteoblast differentiation translates into a lower production of extracellular bone matrix proteins (collagen type I, osteocalcin). Stimulation of the mineralization inhibitors Dmp1 and Phex impairs subsequent matrix mineralization (blue). GCs increase caspase-mediated osteoblast and osteocyte apoptosis through up-regulation of the Bim and Fas/FasL systems. Some osteoprotective drugs, e.g. PTH analogs and bisphosphonates specifically prevent GC-mediated apoptosis. This involves survival pathways steroid receptor coactivator and ERK, Wnt signaling, and gap junctional communication via connexin-43 (green). GCs directly prolong osteoclast survival via caspase 3 but reduce osteoclast activity through blocking macrophage-colony stimulating factor (M-CSF)-mediated pathways. Also, osteoclast number and activity are modulated by enhancing the RANKL/OPG ratio and by inhibiting the proosteoclastic cytokines IL-1, -6, and TNF-α. Denosumab, a human anti-RANKL antibody, is able to prevent GC-induced enhanced osteoclastogenesis (purple). BMP, Bone morphogenetic protein; DKK-1, dickkopf-1; SRC, src proto-oncogene; sFRP-1, secreted frizzled-related protein-1.

Osteoblast fate

The shift toward adipogenesis is mainly mediated by the transactivation of CCAAT/enhancer binding protein δ, which increases the expression of the pivotal adipogenic transcription factor peroxisome proliferator-activated receptor (PPAR)γ2. Concomitantly, the expression of the osteogenic transcription factor runt-related protein 2 (Runx2) is repressed (Fig. 1, orange box) (20). This phenomenon may be recapitulated in patients receiving long-term GC treatment, who concurrently lose bone and accumulate bone marrow fat (21). Further studies have identified additional molecules such as the GC-induced leucine zipper or FHL2 that are activated by GCs to control the expression of PPARγ2 and Runx2 (22, 23). In fact, these molecules are capable of promoting osteogenic differentiation through Runx2 and Wnt signaling (Fig. 1, orange and blue box). Thus, whereas physiological doses of GCs may regulate PPARγ2 and Runx2 in such a way that osteoblast differentiation is favored, pharmacological doses of GCs may preferentially activate PPARγ2 and induce adipogenic differentiation.

Osteoblast maturation and function

Excess GC levels further inhibit osteoblast differentiation as they repress bone-anabolic factors such as bone morphogenetic proteins, IGF-1, and TGFβ, which activate the essential osteoblastic transcription factors Runx2 and β-catenin (Fig. 1, blue box) (15). The canonical Wnt pathway, in particular, is crucial for osteoblastogenesis and mediates its actions via stabilizing cytosolic β-catenin, which then translocates into the nucleus and induces the expression of osteoblast-specific genes. This process is promoted by the destabilization of glycogen synthase kinase 3β (GSK3β), and can be specifically inhibited by secreted proteins such as dickkopf-1 and secreted frizzled-related protein-1. GCs have been shown to suppress Wnt signaling by increasing the expression of these inhibitors and by stabilizing GSK3β (Fig. 1, blue box) (24). However, also in the case of Wnt signaling, GC effects are bivalent, depending on the differentiation state and anatomical site. A recent study has demonstrated that endogenous GCs are in fact necessary for proper skeletal development of the cranium (25). This phenomenon was attributed to decreased canonical Wnt signaling in osteoblasts. Interestingly, long bone development was not affected in these mice, indicating that GCs differentially regulate intramembranous and endochondral bone formation.

In addition to osteoblast differentiation, GCs also impair bone matrix synthesis and composition. Gene expression levels of type I collagen and osteocalcin, two major proteins of the extracellular matrix, are transrepressed by GCs through transcriptional and posttranscriptional mechanisms (15). Moreover, expression levels of mineralization inhibitors such as Dmp-1 and Phex were increased in bone tissue of mice after GC treatment (26). Of note, the osteoporosis drugs, teriparatide, a PTH analog, and the bisphosphonate risedronate, have been shown to improve bone quality in a mouse model of GC-induced osteoporosis partially by counteracting GC effects on mineralization inhibitors (Fig. 1, blue and green box) (26).

Osteoblast and osteocyte apoptosis

Among the various skeletal effects of GCs, the induction of apoptosis of osteoblasts and osteocytes appears to be predominant. This aspect substantially contributes to impaired bone quality because the main function of osteocytes is to sense local microdamage and to induce bone repair. GC-induced apoptosis is associated with enhanced activities of caspases 3, 7, and 8 that are downstream effectors of the activated proapoptotic gene Bim and the Fas/FasL death receptor pathway (Fig. 1, green box) (27, 28, 29). GCs also induce apoptosis in osteoblasts by stabilizing GSK3β activity. Recent studies indicate that bisphosphonates and PTH can reverse the proapoptotic effects of GCs through activating antiapoptotic pathways, including ERK and steroid receptor coactivator as well as Wnt signaling. Interestingly, both drugs enhance osteocyte life span by increasing connexin-43-mediated communication through gap junctions, a critical mechano-sensing pathway (6, 30). Of note, administration of the antiarrhythmic rotigaptide, which modulates gap junctional communication, has been shown to maintain bone mass in the ovariectomized rat, a model of postmenopausal osteoporosis, which is also characterized by enhanced osteoblast/osteocyte apoptosis (31). Thus, modulating cell-cell communication through gap junction and hemichannel formation may represent a novel therapeutic principle.

GC effects on osteoclasts

GCs exert direct and indirect actions on osteoclasts. Although GCs inhibit the expression of proosteoclastic cytokines such as IL-1 and -6 in osteoblasts and stromal cells, the increase of the RANKL/OPG ratio seems to be a key mechanism of GC-induced bone loss (Fig. 1, purple box) (32). Multiple pathways have been proposed to inhibit OPG expression. GCs have been found to transrepress OPG by decreasing the amount of the phospho-c-Jun protein, which maintains steady state transcription of the OPG gene. Promoter activity studies revealed that GC-mediated suppression of OPG production is dependent on canonical Wnt signaling (33). The significance of the RANKL/OPG pathway in the pathogenesis of GC-induced osteoporosis has been underscored in mice expressing a chimeric RANKL protein, which is neutralized by the human monoclonal antibody denosumab. Treatment of these mice with denosumab maintained bone mass and bone strength and suppressed bone resorption during a 4-wk course of GC treatment (34).

The direct effects of GCs on osteoclasts in vitro are ambiguous. In vivo studies using mice overexpressing 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) or lacking the GR specifically in osteoclasts revealed that GCs prolong osteoclast survival by inhibiting caspase 3-dependent apoptosis (Fig. 1, purple box) (35, 36). GCs also reduce osteoclast activity by interfering with the formation of the ruffled border, an organelle that is indispensible for bone resorption. Disruption of the osteoclastic cytoskeleton due to decreased activation of rhoA, rac, and vav3 through macrophage colony-stimulating factor was identified as the underlying mechanism (36). Intriguingly, mice containing the osteoclast-specific knockout of the GR were partially protected from GC-suppressed bone formation, indicating that some of the GC-mediated effects on osteoblasts are indirectly mediated by osteoclasts.

Susceptibility to GC-induced osteoporosis

Interestingly, some individuals are more susceptible to develop GIO than others, indicating that GC bioavailability may vary among individuals. However, predicting the individual GC sensitivity is difficult, because multiple determinants may contribute, including polymorphisms in the GR gene, the activity of the underlying disease, the distribution and metabolism of steroids, the choice of exogenous steroids, the availability and affinity of GRs, the interactions with nuclear coactivators and corepressors, and the presence and local activity of GC-modifying enzymes such as 11β-HSD (37). Especially the 11β-HSD system has emerged as a key regulator of active GC availability because 11β-HSD1 catalyzes the conversion of the inactive cortisone into the active cortisol, and 11β-HSD2 isoform catalyzes the reverse reaction. Of note, exogenously administered GCs differ in their affinities for the 11β-HSD isoforms (38). For example, oxidation of steroids by 11β-HSD2 is diminished in fluorinated (dexamethasone) or methylated (methylprednisolone) GC compounds, and the reduction by 11β-HSD1 depends upon the steroids structure. Thus, it is important to consider the metabolism of synthetic GCs by 11β-HSD as well as the target tissue that may preferentially express 11β-HSD1 or 11β-HSD2 (e.g. colon, kidney) to optimize systemic GC therapy.

Bone-sparing GCs on the horizon?

In the last decade, considerable effort has been made to improve the risk-benefit ratio of GCs. Among these, SEGRAs appear to be the most promising development. By preferring GR-protein interactions rather than GR-DNA binding, these drugs have been found to dissociate transactivating from transrepressing activities. As a result, many compounds have been developed that display similar antiinflammatory actions as conventional GCs but are accompanied with fewer metabolic side effects (Table 1). ZK 216575 and AL-438 are two particular SEGRAs that have been investigated regarding their bone-sparing potential. Although AL-438 was as efficient as prednisolone in reducing inflammation in a rat model of adjuvant arthritis and paw edema, it did not reduce the bone mineral apposition rate or cancellous bone mass compared with prednisolone (39). Also, ZK 216575 has been shown to exert a similar reduction of ear inflammation in rats and mice compared with prednisolone, but a reduced induction of diabetes mellitus and skin atrophy (40). Although the effects on bone were not assessed in this study, in vitro experiments revealed that ZK 216575 may be bone sparing, because it was a poor inducer of RANKL (41). Of note, a recent in vivo study demonstrated that GR dimerization-deficient mice were not protected from GC-induced bone loss (11). This suggests that GC-mediated suppression of bone formation is not entirely dependent on transrepression through GR-DNA binding, but may also depend on protein-protein interactions. Because the structural and functional mechanisms that are required to dissociate transrepression from transactivation to achieve maximal antiinflammatory activity while minimizing side effects are incompletely understood, development of the perfect SEGRA will remain a challenge. Nevertheless, a better understanding of the GR-mediated actions may eventually result in the generation of more specific (nonsteroidal) antiinflammatory ligands.

Table 1.

Profile of SEGRAs

Compound Manufacturer Dissociating effects in vitro Dissociating effects in vivo Reference
AL-438 Abbott-Ligand Yes Yes (39 42 )
BI-115 Boehringer Ingelheim Yes Yes (11 43 )
Compound A University of Ghent Yes Yes (44 45 )
Compound 15 Merck Research Laboratories Yes AI, not tested for SEs (46 )
Compound 25 UC-SF Yes AI, not tested for SEs (47 )
Compound 28 Merck Research Laboratories Yes AI, not tested for SEs (48 )
Compound 60 GSK Yes AI, not tested for SEs (49 )
LGD-5552 Ligand Yes Yes (50 )
RU 24858 Sanofi Aventis Yes No (51 )
ZK 216348 Bayer Schering Pharma Yes Yes (40 )

Modified from Ref. 11 . AI, Antiinflammatory; SEs, side effects.

Conclusion

Long-term treatment with GCs is associated with profound bone loss and increased bone fragility. Across various species and model systems, GCs control the birth (differentiation), life (activity), and death (apoptosis) of bone cells. These fundamental effects translate into insufficient matrix production by osteoblasts, impaired matrix mineralization, increased osteoblast and osteocyte apoptosis, and prolonged life span of osteoclasts, which in sum contribute to bone loss. Some of the molecular signaling cascades underlying these cellular phenomena have been defined and provide novel targets for future therapies such as interfering with the Wnt or RANK pathway. A deeper understanding of the regulation of bone-specific proteins by the GR would facilitate the evaluation of bone-protective SEGRAs and their usefulness in clinical medicine.

Acknowledgments

Owing to space limitations, we have not been able to cite all relevant work; we apologize to those whose work has been omitted.

NURSA Molecule Pages:

  • Ligands: Dexamethasone | Hydrocortisone;

  • Nuclear Receptors: GR.

Footnotes

This work was supported by the European Calcified Tissue Society/Amgen fellowship (to M.R.) and grants from the Center of Regenerative Therapies Dresden of the Technical University Dresden and Grant DFG/Transregio 67, B2 from the Deutsche Forschungsgemeinschaft (to L.C.H.).

Disclosure Summary: The authors have nothing to disclose.

First Published Online May 28, 2009

Abbreviations: GC, Glucocorticoid; GIO, GC-induced osteoperosis; GR, glucocorticoid receptor; GSK3β, glycogen synthase kinase 3β; 11β-HSD2, 11β-hydroxysteroid dehydrogenase 2; OPG, osteoprotegerin; PPAR, peroxisome proliferator-activated receptor; RANKL, receptor activator of nuclear factor-κB ligand; Runx2, runt-related protein 2; SEGRA, selective GR agonist.

References

  • 1.Schäcke H, Döcke WD, Asadullah K2002. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther 96:23–43 [DOI] [PubMed] [Google Scholar]
  • 2.Manolagas SC, Weinstein RS1999. New developments in the pathogenesis and treatment of steroid-induced osteoporosis. J Bone Miner Res 14:1061–1066 [DOI] [PubMed] [Google Scholar]
  • 3.Cooper C, Coupland C, Mitchell M1995. Rheumatoid arthritis, corticosteroid therapy and hip fracture. Ann Rheum Dis 54:49–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Naganathan V, Jones G, Nash P, Nicholson G, Eisman J, Sambrook PN2000. Vertebral fracture risk with long-term corticosteroid therapy: prevalence and relation to age, bone density, and corticosteroid use. Arch Intern Med 160:2917–2922 [DOI] [PubMed] [Google Scholar]
  • 5.Van Staa TP, Laan RF, Barton IP, Cohen S, Reid DM, Cooper C2003. Bone density threshold and other predictors of vertebral fracture in patients receiving oral glucocorticoid therapy. Arthritis Rheum 48:3224–3229 [DOI] [PubMed] [Google Scholar]
  • 6.Cherian PP, Xia X, Jiang JX2008. Role of gap junction, hemichannels, and connexin 43 in mineralizing in response to intermittent and continuous application of parathyroid hormone. Cell Commun Adhes 15:43–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rauner M, Sipos W, Pietschmann P2007. Osteoimmunology. Int Arch Allergy Immunol 143:31–48 [DOI] [PubMed] [Google Scholar]
  • 8.Hofbauer LC, Schoppet M2004. Clinical implications of the osteoprotegerin/RANKL/RANK system for bone and vascular diseases. JAMA 292:490–495 [DOI] [PubMed] [Google Scholar]
  • 9.Song IH, Buttgereit F2006. Non-genomic glucocorticoid effects to provide the basis for new drug developments. Mol Cell Endocrinol 246:142–146 [DOI] [PubMed] [Google Scholar]
  • 10.Beavan S, Horner A, Bord S, Ireland D, Compston J2001. Colocalization of glucocorticoid and mineralocorticoid receptors in human bone. J Bone Miner Res 16:1496–1504 [DOI] [PubMed] [Google Scholar]
  • 11.Schäcke H, Berger M, Rehwinkel H, Asadullah K2007. Selective glucocorticoid receptor agonists (SEGRAs): novel ligands with an improved therapeutic index. Mol Cell Endocrinol 275:109–117 [DOI] [PubMed] [Google Scholar]
  • 12.Lewis-Tuffin LJ, Jewell CM, Bienstock RJ, Collins JB, Cidlowski JA2007. Human glucocorticoid receptor β binds RU-486 and is transcriptionally active. Mol Cell Biol 27:2266–2282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Honda M, Orii F, Ayabe T, Imai S, Ashida T, Obara T, Kohgo Y2000. Expression of glucocorticoid receptor β in lymphocytes of patients with glucocorticoid-resistant ulcerative colitis. Gastroenterology 118:859–866 [DOI] [PubMed] [Google Scholar]
  • 14.Orii F, Ashida T, Nomura M, Maemoto A, Fujiki T, Ayabe T, Imai S, Saitoh Y, Kohgo Y2002. Quantitative analysis for human glucocorticoid receptor α/β mRNA in IBD. Biochem Biophys Res Commun 296:1286–1294 [DOI] [PubMed] [Google Scholar]
  • 15.Canalis E, Mazziotti G, Giustina A, Bilezikian JP2007. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 18:1319–1328 [DOI] [PubMed] [Google Scholar]
  • 16.Dempster DW1989. Bone histomorphometry in glucocorticoid-induced osteoporosis. J Bone Miner Res 4:137–141 [DOI] [PubMed] [Google Scholar]
  • 17.Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC1998. Inhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms of their deleterious effects on bone. J Clin Invest 102:274–282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Eijken M, Hewison M, Cooper MS, de Jong FH, Chiba H, Stewart PM, Uitterlinden AG, Pols HA, van Leeuwen JP2005. 11β-Hydroxysteroid dehydrogenase expression and glucocorticoid synthesis are directed by a molecular switch during osteoblast differentiation. Mol Endocrinol 19:621–631 [DOI] [PubMed] [Google Scholar]
  • 19.Sher LB, Woitge HW, Adams DJ, Gronowicz GA, Krozowski Z, Harrison JR, Kream BE2004. Transgenic expression of 11β-hydroxysteroid dehydrogenase type 2 in osteoblasts reveals an anabolic role for endogenous glucocorticoids in bone. Endocrinology 145:922–929 [DOI] [PubMed] [Google Scholar]
  • 20.Shi XM, Blair HC, Yang X, McDonald JM, Cao X2000. Tandem repeat of C/EBP binding sites mediates PPARγ2 gene transcription in glucocorticoid-induced adipocyte differentiation. J Cell Biochem 76:518–527 [DOI] [PubMed] [Google Scholar]
  • 21.Vande Berg BC, Malghem J, Lecouvet FE, Devogelaer JP, Maldague B, Houssiau FA1999. Fat conversion of femoral marrow in glucocorticoid-treated patients: a cross-sectional and longitudinal study with magnetic resonance imaging. Arthritis Rheum 42:1405–1411 [DOI] [PubMed] [Google Scholar]
  • 22.Zhang W, Yang N, Shi XM2008. Regulation of mesenchymal stem cell osteogenic differentiation by glucocorticoid-induced leucine zipper (GILZ). J Biol Chem 283:4723–4729 [DOI] [PubMed] [Google Scholar]
  • 23.Hamidouche Z, Haÿ E, Vaudin P, Charbord P, Schüle R, Marie PJ, Fromigué O2008. FHL2 mediates dexamethasone-induced mesenchymal cell differentiation into osteoblasts by activating Wnt/β-catenin signaling-dependent Runx2 expression. FASEB J 22:3813–3822 [DOI] [PubMed] [Google Scholar]
  • 24.Wang FS, Ko JY, Yeh DW, Ke HC, Wu HL2008. Modulation of Dickkopf-1 attenuates glucocorticoid induction of osteoblast apoptosis, adipocytic differentiation, and bone mass loss. Endocrinology 149:1793–1801 [DOI] [PubMed] [Google Scholar]
  • 25.Zhou H, Mak W, Kalak R, Street J, Fong-Yee C, Zheng Y, Dunstan CR, Seibel MJ2009. Glucocorticoid-dependent Wnt signaling by mature osteoblasts is a key regulator of cranial skeletal development in mice. Development 136:427–436 [DOI] [PubMed] [Google Scholar]
  • 26.Yao W, Cheng Z, Pham A, Busse C, Zimmermann EA, Ritchie RO, Lane NE2008. Glucocorticoid-induced bone loss in mice can be reversed by the actions of parathyroid hormone and risedronate on different pathways for bone formation and mineralization. Arthritis Rheum 58:3485–3497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kogianni G, Mann V, Ebetino F, Nuttall M, Nijweide P, Simpson H, Noble B2004. Fas/CD95 is associated with glucocorticoid-induced osteocyte apoptosis. Life Sci 75:2879–2895 [DOI] [PubMed] [Google Scholar]
  • 28.O'Brien CA, Jia D, Plotkin LI, Bellido T, Powers CC, Stewart SA, Manolagas SC, Weinstein RS2004. Glucocorticoids act directly on osteoblasts and osteocytes to induce their apoptosis and reduce bone formation and strength. Endocrinology 145:1835–1841 [DOI] [PubMed] [Google Scholar]
  • 29.Espina B, Liang M, Russell RG, Hulley PA2008. Regulation of bim in glucocorticoid-mediated osteoblast apoptosis. J Cell Physiol 215:488–496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Plotkin LI, Lezcano V, Thostenson J, Weinstein RS, Manolagas SC, Bellido T2008. Connexin 43 is required for the anti-apoptotic effect of bisphosphonates on osteocytes and osteoblasts in vivo. J Bone Miner Res 23:1712–1721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jørgensen NR, Teilmann SC, Henriksen Z, Meier E, Hansen SS, Jensen JE, Sørensen OH, Petersen JS2005. The antiarrhythmic peptide analog rotigaptide (ZP123) stimulates gap junction intercellular communication in human osteoblasts and prevents decrease in femoral trabecular bone strength in ovariectomized rats. Endocrinology 146:4745–4754 [DOI] [PubMed] [Google Scholar]
  • 32.Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, Spelsberg TC, Khosla S1999. Stimulation of osteoprotegerin ligand and inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage cells: potential paracrine mechanisms of glucocorticoid-induced osteoporosis. Endocrinology 140:4382–4389 [DOI] [PubMed] [Google Scholar]
  • 33.Kondo T, Kitazawa R, Yamaguchi A, Kitazawa S2008. Dexamethasone promotes osteoclastogenesis by inhibiting osteoprotegerin through multiple levels. J Cell Biochem 103:335–345 [DOI] [PubMed] [Google Scholar]
  • 34.Hofbauer LC, Zeitz U, Schoppet M, Skalicky M, Schüler C, Stolina M, Kostenuik PJ, Erben RG2009. Prevention of glucocorticoid-induced bone loss in mice by inhibition of RANKL. Arthritis Rheum 60:1427–1437 [DOI] [PubMed] [Google Scholar]
  • 35.Jia D, O'Brien CA, Stewart SA, Manolagas SC, Weinstein RS2006. Glucocorticoids act directly on osteoclasts to increase their life span and reduce bone density. Endocrinology 147:5592–5599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, Muglia LJ, Ross FP, Teitelbaum SL2006. Glucocorticoids suppress bone formation via the osteoclast. J Clin Invest 116:2152–2160 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tomlinson JW, Walker EA, Bujalska IJ, Draper N, Lavery GG, Cooper MS, Hewison M, Stewart PM2004. 11β-Hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Endocr Rev 25:831–866 [DOI] [PubMed] [Google Scholar]
  • 38.Diederich S, Eigendorff E, Burkhardt P, Quinkler M, Bumke-Vogt C, Rochel M, Seidelmann D, Esperling P, Oelkers W, Bähr V2002. 11β-Hydroxysteroid dehydrogenase types 1 and 2: an important pharmacokinetic determinant for the activity of synthetic mineralo- and glucocorticoids. J Clin Endocrinol Metab 87:5695–5701 [DOI] [PubMed] [Google Scholar]
  • 39.Coghlan MJ, Jacobson PB, Lane B, Nakane M, Lin CW, Elmore SW, Kym PR, Luly JR, Carter GW, Turner R, Tyree CM, Hu J, Elgort M, Rosen J, Miner JN2003. A novel antiinflammatory maintains glucocorticoid efficacy with reduced side effects. Mol Endocrinol 17:860–869 [DOI] [PubMed] [Google Scholar]
  • 40.Schäcke H, Schottelius A, Döcke WD, Strehlke P, Jaroch S, Schmees N, Rehwinkel H, Hennekes H, Asadullah K2004. Dissociation of transactivation from transrepression by a selective glucocorticoid receptor agonist leads to separation of therapeutic effects from side effects. Proc Natl Acad Sci USA 101:227–232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Humphrey EL, Williams JH, Davie MW, Marshall MJ2006. Effects of dissociated glucocorticoids on OPG and RANKL in osteoblastic cells. Bone 38:652–661 [DOI] [PubMed] [Google Scholar]
  • 42.Owen HC, Miner JN, Ahmed SF, Farquharson C2007. The growth plate sparing effects of the selective glucocorticoid receptor modulator, AL-438. Mol Cell Endocrinol 264:164–170 [DOI] [PubMed] [Google Scholar]
  • 43.Kuzmich D, Kirrane T, Proudfoot J, Bekkali Y, Zindell R, Beck L, Nelson R, Shih CK, Kukulka AJ, Paw Z, Reilly P, Deleon R, Cardozo M, Nabozny G, Thomson D2007. Identification of dissociated non-steroidal glucocorticoid receptor agonists. Bioorg Med Chem Lett 17:5025–5031 [DOI] [PubMed] [Google Scholar]
  • 44.Dewint P, Gossye V, De Bosscher K, Vanden Berghe W, Van Beneden K, Deforce D, Van Calenbergh S, Müller-Ladner U, Vander Cruyssen B, Verbruggen G, Haegeman G, Elewaut D2008. A plant-derived ligand favoring monomeric glucocorticoid receptor conformation with impaired transactivation potential attenuates collagen-induced arthritis. J Immunol 180:2608–2615 [DOI] [PubMed] [Google Scholar]
  • 45.De Bosscher K, Vanden Berghe W, Beck IM, Van Molle W, Hennuyer N, Hapgood J, Libert C, Staels B, Louw A, Haegeman G2005. A fully dissociated compound of plant origin for inflammatory gene repression. Proc Natl Acad Sci USA 102:15827–15832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Thompson CF, Quraishi N, Ali A, Tata JR, Hammond ML, Balkovec JM, Einstein M, Ge L, Harris G, Kelly TM, Mazur P, Pandit S, Santoro J, Sitlani A, Wang C, Williamson J, Miller DK, Yamin TT, Thompson CM, O'Neill EA, Zaller D, Forrest MJ, Carballo-Jane E, Luell S2005. Novel heterocyclic glucocorticoids: in vitro profile and in vivo efficacy. Bioorg Med Chem Lett 15:2163–2167 [DOI] [PubMed] [Google Scholar]
  • 47.Shah N, Scanlan TS2004. Design and evaluation of novel nonsteroidal dissociating glucocorticoid receptor ligands. Bioorg Med Chem Lett 14:5199–5203 [DOI] [PubMed] [Google Scholar]
  • 48.Smith CJ, Ali A, Balkovec JM, Graham DW, Hammond ML, Patel GF, Rouen GP, Smith SK, Tata JR, Einstein M, Ge L, Harris GS, Kelly TM, Mazur P, Thompson CM, Wang CF, Williamson JM, Miller DK, Pandit S, Santoro JC, Sitlani A, Yamin TT, O'Neill EA, Zaller DM, Carballo-Jane E, et al.2005. Novel ketal ligands for the glucocorticoid receptor: in vitro and in vivo activity. Bioorg Med Chem Lett 15:2926–2931 [DOI] [PubMed] [Google Scholar]
  • 49.Barker M, Clackers M, Copley R, Demaine DA, Humphreys D, Inglis GG, Johnston MJ, Jones HT, Haase MV, House D, Loiseau R, Nisbet L, Pacquet F, Skone PA, Shanahan SE, Tape D, Vinader VM, Washington M, Uings I, Upton R, McLay IM, Macdonald SJ2006. Dissociated nonsteroidal glucocorticoid receptor modulators; discovery of the agonist trigger in a tetrahydronaphthalene-benzoxazine series. J Med Chem 49:4216–4231 [DOI] [PubMed] [Google Scholar]
  • 50.López FJ, Ardecky RJ, Bebo B, Benbatoul K, De Grandpre L, Liu S, Leibowitz MD, Marschke K, Rosen J, Rungta D, Viveros HO, Yen WC, Zhi L, Negro-Vilar A, Miner JN2008. LGD-5552, an antiinflammatory glucocorticoid receptor ligand with reduced side effects, in vivo Endocrinology 149:2080–2089 [DOI] [PubMed] [Google Scholar]
  • 51.Belvisi MG, Wicks SL, Battram CH, Bottoms SE, Redford JE, Woodman P, Brown TJ, Webber SE, Foster ML2001. Therapeutic benefit of a dissociated glucocorticoid and the relevance of in vitro separation of transrepression from transactivation activity. J Immunol 166:1975–1982 [DOI] [PubMed] [Google Scholar]

Articles from Molecular Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES