REVIEW
Mechanisms modulating inflammatory osteolysis: A review with insights into therapeutic targets

https://doi.org/10.1016/j.prp.2008.07.002Get rights and content

Abstract

Inflammatory osteolysis is a relatively frequent and incapacitating complication of rheumatoid arthritis and multiple other inflammation-associated bone diseases. It is thought to operate through an ultimate common pathway of accelerated osteoclast recruitment and activation under the control of cytokines produced in the inflammatory environment. Over the past decade, there have been major advances in our understanding of the mechanisms of osteoclastogenesis. It is now clear that the interaction of receptor activator NF-κB (RANK) and its ligand, RANKL, plays a central role in osteoclast formation and activity. Therefore, understanding osteoclastogenesis offers new pathways for potential therapeutic intervention in inflammatory osteolysis. The success of anti-tumor necrosis factor-α and interleukin-1 therapy highlights the central role that these specific cytokines play in this disease. This review outlines our current understanding of the mechanisms mediating inflammatory osteolysis and highlights potential therapeutic strategies.

Introduction

Destructive erosion of bone or osteolysis is a major complication of inflammatory conditions such as rheumatoid arthritis (RA), periodontal disease, and periprosthetic osteolysis. RA is an autoimmune disease that affects approximately 1.0% of US adults, with a female to male ratio of 2.5 to 1 [52]. Its hallmark is progressive joint destruction which causes major morbidity. The etiology of RA is largely unclear. The combination of genetic susceptibility with environmental factors is considered to play a role in the initiation of an immunologic response against the synovium. Periodontal disease is highly prevalent and can affect up to 90% of the world's population. It is well known as the leading cause of tooth loss in adults [72]. Despite its prevalence, little is known about the mechanism by which periodontal bone erosion occurs, although host response to pathogenic microorganisms present in the mouth appears to trigger the process. In addition, genetic and environmental factors are also thought to contribute to the cause of this disease [72]. Periprosthetic osteolysis is caused by chronic bone resorption around exogenous implant devices until fixation is lost [28], and is considered as resulting from an innate immune response to wear-debris particles, with little contribution by components of the acquired immune system [22].

Although these conditions are initiated by distinct causes and progress by alternative pathways, the important common factor(s) in the pathological process of these diseases are over-production of proinflammatory cytokines and excessive destruction of bone by osteoclasts near the site of inflammation. The bone erosion seen in these conditions is largely localized to the inflamed tissues, distinct from systemic, hormonally regulated bone pathologies, such as osteoporosis. These inflamed tissues, found in many of these diseases, also produce proinflammatory cytokines, i.e., TNF-α, IL-1, and IL-6, that are, in turn, involved in osteoclast differentiation signaling and bone-resorbing activities. Thus, inflammatory osteolysis is the product of enhanced osteoclast recruitment and activation prompted by proinflammatory cytokines. The osteoclast, which is the sole bone-resorbing cell, is therefore central to the pathogenesis of inflammatory osteolysis. Understanding the mechanisms by which osteoclasts resorb bone and the cytokines that regulate their differentiation and activity provides mechanism-based potential therapeutic targets to prevent inflammatory bone loss.

Section snippets

Osteoclastogenesis in physiological conditions

Osteoclasts are multinucleated cells formed by the fusion of mononuclear precursors of the monocyte/macrophage lineage under the influence of the specific osteoclastogenic cytokine, receptor activator NF-κB ligand (RANKL), and macrophage colony-stimulating factor (M-CSF) [93]. Mice genetically deficient in RANKL [12] or M-CSF [109] signaling do not form osteoclasts and thus develop the pathologic condition, osteopetrosis. RANKL, a member of the TNF superfamily, is a membrane-bound homotrimeric

Osteoclastogenesis in inflammatory conditions

In states of juxtaskeletal inflammation such as RA, the osteoclastogenic molecule RANKL is also produced in abundance by activated T lymphocytes and synovial fibroblasts [24], [45], whether they are in joint or bone. RANKL may also be cleaved from the cell membrane and then interact with RANK as a soluble ligand. Additionally, these same activated synovial fibroblasts also express M-CSF and OPG [44]. Subsequently, blocking of RANKL with OPG at the onset of induced RA in mice prevents bone and

Current and potential future therapeutic targets for inflammatory osteolysis

The recent advances in basic research in understanding the mechanisms of inflammatory osteolysis lead to two major treatment approaches: anti-inflammatory agents and bone resorption-blocking agents.

Conclusions

Patients with RA and other inflammatory bone diseases face complications of joint destruction. The differentiation and function of osteoclasts, the culprits in these closely associated conditions, are accelerated under the control of cytokines produced in the inflammatory environment, in which TNF-α, IL-1, and RANKL play a central role. Understanding this complex process has been helpful in the clinical translation from mechanism to drug development. The approach to combine anti-inflammatory

References (112)

  • S.R. Paccani et al.

    Nonsteroidal anti-inflammatory drugs suppress T-cell activation by inhibiting p38 MAPK induction

    J. Biol. Chem.

    (2002)
  • B.L. Pihlstrom et al.

    Periodontal diseases

    Lancet

    (2005)
  • S.E. Raper et al.

    Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer

    Mol. Genet. Metab.

    (2003)
  • E. Romas et al.

    Involvement of receptor activator of NFkappaB ligand and tumor necrosis factor-alpha in bone destruction in rheumatoid arthritis

    Bone

    (2002)
  • R. Seibl et al.

    Expression and regulation of Toll-like receptor 2 in rheumatoid arthritis synovium

    Am. J. Pathol.

    (2003)
  • S.M. Sharma et al.

    MITF and PU.1 recruit p38 MAPK and NFATc1 to target genes during osteoclast differentiation

    J. Biol. Chem.

    (2007)
  • W.S. Simonet et al.

    Osteoprotegerin: a novel secreted protein involved in the regulation of bone density

    Cell

    (1997)
  • M. Suzuki et al.

    The role of p38 mitogen-activated protein kinase in IL-6 and IL-8 production from the TNF-alpha- or IL-1beta-stimulated rheumatoid synovial fibroblasts

    FEBS Lett.

    (2000)
  • S.L. Teitelbaum

    Osteoclasts: what do they do and how do they do it?

    Am. J. Pathol.

    (2007)
  • D. Vanags et al.

    Therapeutic efficacy and safety of chaperonin 10 in patients with rheumatoid arthritis: a double-blind randomised trial

    Lancet

    (2006)
  • J. Adriaansen et al.

    Gene therapy as a therapeutic approach for the treatment of rheumatoid arthritis: innovative vectors and therapeutic genes

    Rheumatology, Oxford

    (2006)
  • E. Andreakos et al.

    Is targeting Toll-like receptors and their signaling pathway a useful therapeutic approach to modulating cytokine-driven inflammation?

    Immunol. Rev.

    (2004)
  • W.P. Arend

    The mode of action of cytokine inhibitors

    J. Rheumatol. Suppl.

    (2002)
  • R. Beyaert et al.

    The p38/RK mitogen-activated protein kinase pathway regulates interleukin-6 synthesis response to tumor necrosis factor

    Embo J.

    (1996)
  • J.J. Body et al.

    A phase I study of AMGN-0007, a recombinant osteoprotegerin construct, in patients with multiple myeloma or breast carcinoma related bone metastases

    Cancer

    (2003)
  • M. Cella et al.

    Impaired differentiation of osteoclasts in TREM-2-deficient individuals

    J. Exp. Med.

    (2003)
  • L.M. Childs et al.

    In vivo RANK signaling blockade using the receptor activator of NF-kappaB:Fc effectively prevents and ameliorates wear debris-induced osteolysis via osteoclast depletion without inhibiting osteogenesis

    J. Bone Miner. Res.

    (2002)
  • J.M. Dayer

    Interleukin 1 or tumor necrosis factor-alpha: which is the real target in rheumatoid arthritis?

    J. Rheumatol. Suppl.

    (2002)
  • W.C. Dougall et al.

    RANK is essential for osteoclast and lymph node development

    Genes Dev.

    (1999)
  • J.C. Edwards et al.

    Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis

    N. Engl. J. Med.

    (2004)
  • P. Emery

    The therapeutic potential of costimulatory blockade with CTLA4Ig in rheumatoid arthritis

    Expert Opin. Invest. Drugs

    (2003)
  • P. Emery et al.

    The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial

    Arthritis Rheum.

    (2006)
  • M. Feldmann et al.

    Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned?

    Annu. Rev. Immunol.

    (2001)
  • G. Franzoso et al.

    Requirement for NF-kappaB in osteoclast and B-cell development

    Genes Dev.

    (1997)
  • D.E. Furst et al.

    Updated consensus statement on biological agents for the treatment of rheumatic diseases, 2006

    Ann Rheum. Dis.

    (2006)
  • M.C. Genovese et al.

    Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition

    N. Engl. J. Med.

    (2005)
  • F. Goldblatt et al.

    New therapies for rheumatoid arthritis

    Clin. Exp. Immunol.

    (2005)
  • F. Goldblatt et al.

    New therapies for systemic lupus erythematosus

    Clin. Exp. Immunol.

    (2005)
  • S.R. Goldring et al.

    Formation of a synovial-like membrane at the bone–cement interface. Its role in bone resorption and implant loosening after total hip replacement

    Arthritis Rheum.

    (1986)
  • J.A. Goya et al.

    Effect of topical administration of monosodium olpadronate on experimental periodontitis in rats

    J. Periodontol.

    (2006)
  • E.M. Gravallese et al.

    Synovial tissue in rheumatoid arthritis is a source of osteoclast differentiation factor

    Arthritis Rheum.

    (2000)
  • E.M. Gravallese

    Bone destruction in arthritis

    Ann. Rheum. Dis.

    (2002)
  • A.E. Grigoriadis et al.

    c-Fos: a key regulator of osteoclast-macrophage lineage determination and bone remodeling

    Science

    (1994)
  • J.F. Harms et al.

    A small molecule antagonist of the alpha(v)beta3 integrin suppresses MDA-MB-435 skeletal metastasis

    Clin. Exp. Metastasis

    (2004)
  • W.H. Harris

    The problem is osteolysis

    Clin. Orthop. Relat. Res.

    (1995)
  • L.C. Hofbauer et al.

    Role of receptor activator of nuclear factor-kappaB ligand and osteoprotegerin in bone cell biology

    J. Mol. Med.

    (2001)
  • P.G. Hogan et al.

    Transcriptional regulation by calcium, calcineurin, and NFAT

    Genes Dev.

    (2003)
  • V. Iotsova et al.

    Osteopetrosis in mice lacking NF-kappaB1 and NF-kappaB2

    Nat. Med.

    (1997)
  • C. Jorgensen et al.

    Stem cells for repair of cartilage and bone: the next challenge in osteoarthritis and rheumatoid arthritis

    Ann. Rheum. Dis.

    (2001)
  • J. Kaiser

    Gene therapy. Seeking the cause of induced leukemias in X-SCID trial

    Science

    (2003)
  • Cited by (35)

    • 6-gingerol: A therapeutically potent lead candidate in anti-inflammatory drug discovery

      2019, Discovery and Development of Anti-inflammatory Agents from Natural Products
    • Swertiamarin, a natural steroid, prevent bone erosion by modulating RANKL/RANK/OPG signaling

      2017, International Immunopharmacology
      Citation Excerpt :

      The dysfunction of cytokines induces the differentiation and activation of osteoclasts to involve in joint destruction in RA. Inhibition of osteoclastogenesis is always a potential therapeutic strategy for arthritis [41]. The osteoclast cells were identified using multinucleated appearances, tartrate resistant acid phosphatase (TRAP) staining, expression of calcitonin receptors in cell surface and absorb calcified matrices into the cells [42].

    • Mesenchymal Stem Cells as Regulators of the Bone Marrow and Bone Components

      2016, Mesenchymal Stromal Cells as Tumor Stromal Modulators
    • Adsorbed fibrinogen leads to improved bone regeneration and correlates with differences in the systemic immune response

      2013, Acta Biomaterialia
      Citation Excerpt :

      These apparently contradictory data support the idea that the effect of TNF-α is time- or dose-dependent [12]. Also, osteolysis is modulated by inflammation, since differentiation of monocyte/macrophage lineage precursors into osteoclasts is controlled by macrophage colony-stimulating factor (M-CSF) [14] and by receptor of NF-KB ligand (RANKL), which can be produced by osteoblasts, activated T lymphocytes and NK cells [15]. In this study we have used a fibrinogen-coated biomaterial to explore the role of inflammation in bone regeneration.

    • Gingival lymphatic drainage protects against porphyromonas gingivalis-induced bone loss in mice

      2012, American Journal of Pathology
      Citation Excerpt :

      The accumulated neutrophils close to the alveolar bone may degranulate and release superoxide anion resulting in degradation of soft tissue and bone in line with what has been described in the pathogenesis of inflammatory arthritis.26 B cells in periodontal lesions are also linked to periodontal disease development together with T cells, as they contribute substantially to production of receptor activator of nuclear factor-κB ligand (RANKL),27 a cytokine known to induce osteoclast activity28 (see below). Furthermore, the K14 infected mice had significantly higher levels of the proinflammatory cytokines IFN-γ and IL-1β.

    View all citing articles on Scopus

    Supported by NIH grants CA93796, CA098543, AR046031, and the Haley's Hope Memorial Support Fund for Osteosarcoma Research at the University of Alabama at Birmingham, Birmingham, Alabama.

    View full text