Abstract

Periprosthetic osteolysis induced by orthopedic implant-wear particles continues to be the leading cause of arthroplasty failure in majority of patients. Release of the wear debris results in a chronic local inflammatory response typified by the recruitment of immune cells, including macrophages. The cellular mediators derived from activated macrophages favor the osteoclast-bone resorbing activity resulting in bone loss at the site of implant and loosening of the prosthetic components. Emerging evidence suggests that chemokines and their receptors are involved in the progression of periprosthetic osteolysis associated with aseptic implant loosening. In the current study, we investigated the potential role of chemokine C-motif-ligand-1 (XCL1) in the pathogenesis of inflammatory osteolysis induced by wear particles. Expressions of XCL1 and its receptor XCR1 were evident in synovial fluids and tissues surrounding hip-implants of patients undergoing revision total hip arthroplasty. Furthermore, murine calvarial osteolysis model induced by ultra-high molecular weight polyethylene (UHMWPE) particles was used to study the role of XCL1 in the development of inflammatory osteolysis. Mice received single injection of recombinant XCL1 onto the calvariae after implantation of particles exhibited significantly greater osteolytic lesions than the control mice. In contrast, blockade of XCL1 by neutralizing antibody significantly reduced bone erosion and the number of bone-resorbing mature osteoclasts induced by UHMWPE particles. In consistence with the results, transplantation of XCL1-soaked sponge onto calvariae caused osteolytic lesions coincident with excessive infiltration of inflammatory cells and osteoclasts. These results suggested that XCL1 might be involved in the development of periprosthetic osteolysis through promoting infiltration of inflammatory cells and bone resorbing-osteoclasts. Our further results demonstrated that supplementing recombinant XCL1 to cultured human monocytes stimulated with the receptor activator of nuclear factor kappa-B ligand (RANKL) promoted osteoclastogenesis and the osteoclast-bone resorbing activity. Moreover, recombinant XCL1 promoted the expression of inflammatory and osteoclastogenic factors, including IL-6, IL-8, and RANKL in human differentiated osteoblasts. Together, these results suggested the potential role of XCL1 in the pathogenesis of periprosthetic osteolysis and aseptic loosening. Our data broaden knowledge of the pathogenesis of aseptic prosthesis loosening and highlight a novel molecular target for therapeutic intervention.

Highlights

  • Total joint arthroplasty (TJA) is one of the most successful medical procedures in modern clinical orthopedics, which restores the mobility of patients in the late-stage arthritis

  • It is significant to note that majority of XCR1-positive cells were co-stained with the F4/80, CD68, and iNOS antibodies used as macrophages/dendritic cells markers (Figures 1A–C)

  • Bearing in mind that fibroblasts are the dominant cell type in the interface membrane and play a role in the pathogenesis of periprosthetic osteolysis associated with particulate wear debris [15], we examined the effects of recombinant X-C motif chemokine ligand 1 (XCL1) on the expression of inflammatory and osteoclastogenesis factors in Human Fibroblast-Like Synoviocytes (HFLS)

Read more

Summary

Introduction

Total joint arthroplasty (TJA) is one of the most successful medical procedures in modern clinical orthopedics, which restores the mobility of patients in the late-stage arthritis. An increase in the gene expressions of chemokines such as CCL2, CCL3, and CXCL8 is evident in periprosthetic tissues surrounding aseptically loosening implants [2] In line with this view, blocking CCL2, CXCL2, CXCL12, and their receptors has shown substantial reduction in the osteolytic activity in a murine osteolysis model, suggesting that they play crucial roles in the progression of osteolysis. These findings highlight that the interaction of chemokine-chemokine receptors is a promising therapeutic target for preventing implant loosening [2, 5, 6]. Identifying of chemokines involved in the pathogenesis of periprosthetic osteolysis may provide a clue for development of new therapeutics

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

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