Abstract

BackgroundGene therapy has the potential to provide long-term production of therapeutic proteins in the joints of osteoarthritis (OA) patients. The objective of this study was to analyse the therapeutic potential of disease-inducible expression of anti-inflammatory interleukin-10 (IL-10) in the three-dimensional micromass model of the human synovial membrane.MethodsSynovial tissue samples from OA patients were digested and the cells were mixed with Matrigel to obtain 3D micromasses. The CXCL10 promoter combined with the firefly luciferase reporter in a lentiviral vector was used to determine the response of the CXCL10 promoter to tumour necrosis factor alpha (TNF-α), interleukin-1β (IL-1β) and lipopolysaccharide (LPS). The effects of recombinant IL-10 on gene expression were determined by quantitative PCR. The production of IL-10 from the CXCL10p-IL10 vector and the effects on pro-inflammatory cytokine production were assessed by multiplex ELISA.ResultsMicromasses made from whole synovial membrane cell suspensions form a distinct surface composition containing macrophage and fibroblast-like synoviocytes thus mimicking the synovial lining. This lining can be transduced by lentiviruses and allow CXCL-10 promoter-regulated transgene expression. Adequate amounts of IL-10 transgene were produced after stimulation with pro-inflammatory factors able to reduce the production of synovial IL-1β and IL-6.ConclusionsSynovial micromasses are a suitable model to test disease-regulated gene therapy approaches and the CXCL10p-IL10 vector might be a good candidate to decrease the inflammatory response implicated in the pathogenesis of OA.Electronic supplementary materialThe online version of this article (doi:10.1186/s13075-016-1083-1) contains supplementary material, which is available to authorized users.

Highlights

  • Gene therapy has the potential to provide long-term production of therapeutic proteins in the joints of osteoarthritis (OA) patients

  • Sustained culture of fibroblast-like synoviocytes (FLS) can result in phenotype alterations [20] and in FLS micromasses the contribution of macrophage-like synoviocytes (MLS) is omitted

  • The sections were stained using the 11-fibrau antibody, which revealed that most cells in the lining are synovial fibroblast-like cells (Fig. 1b)

Read more

Summary

Introduction

Gene therapy has the potential to provide long-term production of therapeutic proteins in the joints of osteoarthritis (OA) patients. Interleukin-1β (IL-1β) and tumour necrosis factor alpha (TNF-α) are key pro-inflammatory cytokines, primarily produced by macrophages that infiltrate the synovium during inflammatory OA [3] These cytokines trigger the release of other cytokines and matrix-degrading enzymes from resident articular cells, including the fibroblast-like synoviocytes (FLS) that damage the articular cartilage. Intraarticular injections are feasible but proteins injected in the joint are rapidly cleared and the invasive injections have to be repeated increasing the discomfort for patients [6]. These considerations ask for the development of a different strategy to obtain long-term drug effects in the OA joint. Local gene therapy can be a suitable approach and might even be used to express proteins with a short half-life like interleukin-10 (IL-10), which is a potent anti-inflammatory cytokine [7]

Objectives
Methods
Results
Discussion
Conclusion
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.