Abstract

Approximately 30 members of the tumor necrosis factor receptor superfamily (TNFRSF) have been identified. They are transmembrane proteins with cysteine‐rich motifs in their extracellular domains that bind to their cognate ligands 1. They are categorized into 3 groups: death domain–containing receptors, decoy receptors, and TNFR‐associated factor–binding receptors. Only 8 TNFRSF members contain a death domain (TNFR type I [TNFRI], death receptor 3 [DR‐3], DR‐4, DR‐5, DR‐6, Fas, nerve growth factor receptor, and ectodysplasin A receptor [EDAR]), of which TNFRI and DR‐3 constitute the principal focus of this article. Interactions between TNF superfamily (TNFSF) ligands and TNFRSF receptors help maintain tissue homeostasis by controlling survival, proliferation, differentiation, and effector function of immune cells. We limit our review to recent advances and novel insights into the roles of TNFRI and DR‐3 in bone and joint biology. Bone cells (osteoblasts, osteoclasts, and osteocytes), fibroblast‐like synoviocytes, chondrocytes, and immune cells that infiltrate the arthritic joint will at different times express a wide range of TNFRSF members and TNFSF ligands. An overview of the current status of our knowledge in this regard is provided in Table 1. The impact of TNFRI activation on bone and inflammatory joint diseases has been researched in great depth 2, 3, but little or no data in the field have been reported on other more recently discovered TNFRSF members such as TROY (TNFRSF expressed on the mouse embryo; TNFRSF19), EDAR, and XEDAR (X‐linked ectodysplasin receptor; TNFRSF27). The unexpected interaction between progranulin (PGRN) and both TNFRI and TNFRII is particularly interesting in the context of arthritis‐associated bone pathology. PGRN levels are elevated in the synovial fluid of patients with rheumatoid arthritis (RA), osteoarthritis (OA), and other arthropathies 4, 5, 6, and PGRN has been shown to inhibit TNF‐induced osteoclastogenesis and promote osteoblast differentiation in mice 7. However, PGRN has a higher binding affinity for TNFRII (antiinflammatory with osteoprotective function) than for TNFRI (predominantly proinflammatory with degenerative function), which suggests conflicting actions. The potential overall impact of these divergent PGRN signaling pathways on the architecture of the arthritic joint has been evaluated 8. Table 1 Cellular expression of death domain–containing TNFRSF members and their association with arthritis*

Highlights

  • About 30 members of tumor necrosis factor receptor superfamily (TNFRSF) have been identified. They are transmembrane proteins with cysteine-rich motifs in their extracellular domains that bind to their cognate ligands [1]. They are categorized into three groups; death domain-containing receptors, decoy receptors, and TNF receptor-associated factor-binding receptors

  • Eight TNFRSF members contain a death domain (TNFR1, Death receptor 3 (DR3), DR4, DR5, DR6, Fas, NGFR, EDAR) of which TNFR1 and DR3 constitute the principle focus of this article

  • This review provides further insight to DR3’s role in bone remodeling and arthritis

Read more

Summary

Introduction

About 30 members of tumor necrosis factor receptor superfamily (TNFRSF) have been identified. The associations of PGRN with TNFR and DR3 led to investigations on the immunological mechanisms underlying PGRN mediated anti-inflammatory and protective activities in autoimmune diseases[67,68,69] Since both animal and human studies have demonstrated that regulatory T cells (Tregs) play a critical role in the prevention of autoimmunity and other pathological immune responses, the effects of PGRN on Treg differentiation and function were first determined. The additional specific inhibition of the TL1A/DR3 interaction and activation of TNFR2 anti-inflammatory pathway by PGRN or its derivate [66] is a unique characteristic and might represent a significant advantage over conventional TNFα inhibitors for patients with refractory or relapsing disease under conventional TNFα-blockers. Looking at the underlying theory and the known preclinical data, Atsttrin could be a therapeutic alternative in cases of refractory or recurrent arthritis

16. Liu CJ
26. Aggarwal BB: Editorial
36. Wang EC
# Key points
Findings
Clinical Perspective
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.