Article Text

Download PDFPDF

AB0090 N-ACETYL-L-CYSTEINE (NAC) controls osteoclastogenesis through regulating TH17 differentiation and RANKL production in rheumatoid arthritis
Free
  1. H-R Kim,
  2. K-A Lee,
  3. S-H Lee
  1. Rheumatology, Konkuk University School of Medicine, Seoul, Korea, Republic Of

Abstract

Background NAC is a thiolic antioxidant produced by the body and serves as a precursor of glutathione synthesis. In rheumatoid arthritis (RA), oxidative stress is an important mechanism causing destructive proliferative synovitis.

Objectives This study aimed to determine the regulatory role of N-Acetyl-L-cysteine (NAC), an antioxidant, in IL-17-induced osteoclast differentiation in RA.

Methods After RA synovial fibroblasts were stimulated by IL-17, the expression and production of RANKL was determined by real-time PCR and ELISA. Human peripheral blood monocytes were cultured with M-CSF, IL-17, RANKL, and/or various concentrations of NAC, followed by counting of the cells for tartrate-resistant acid phosphatase activity to determine osteoclast formation. Osteoclastogenesis was also determined after cocultures of IL-17-stimulated RA synovial fibroblasts, Th17 cells and various concentrations of NAC with monocytes. After human peripheral CD4+ T cells were cultured with NAC under Th17 condition, IL-17, IFN-g, IL-4, Foxp3, RANKL and IL-2 expression and production was determined by flow cytometry or ELISA.

Results When RA synovial fibroblasts were stimulated by IL-17, IL-17 stimulated the production of RANKL, and NAC reduced the IL-17-induced RANKL production in a dose-dependent manner. NAC decreased IL-17-activated phosphorylation of mTOR, JNK and IkB. When human peripheral blood CD14+ monocytes were cultured with M-CSF and IL-17 or RANKL, osteoclasts were differentiated, and NAC reduced the osteoclastogenesis. After human peripheral CD4+ T cells were co-cultured with IL-17-pretreated RA synovial fibroblasts or Th17 cells, NAC reduced their osteoclastogenesis. Under Th17 polarizing condition, NAC decreased Th17 cell differentiation and IL-17 and RANKL production.

Conclusions NAC inhibits the IL-17-induced RANKL production in RA synovial fibroblasts and IL-17-induced osteoclast differentiation. NAC also reduced Th17 polarization. NAC could be a supplementary therapeutic option for inflammatory and bony destructive processes in RA.

Acknowledgements This research was supported by a grant of the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology, Republic of Korea (NRF-2014R1A2A2A01007223) and the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI13C1704).

Disclosure of Interest None declared

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.