Abstract

Background: Calcium pyrophosphate (CPP) microcrystal deposition is associated with wide clinical phenotypes, including acute and chronic arthritis, that are interleukin 1β (IL-1β)-driven. Two CPP microcrystals, namely monoclinic and triclinic CPP dihydrates (m- and t-CPPD), have been identified in human tissues in different proportions according to clinical features. m-CPP tetrahydrate beta (m-CPPTβ) and amorphous CPP (a-CPP) phases are considered as m- and t-CPPD crystal precursors in vitro.Objectives: We aimed to decipher the inflammatory properties of the three crystalline phases and one amorphous CPP phase and the intracellular pathways involved.Methods: The four synthesized CPP phases and monosodium urate crystals (MSU, as a control) were used in vitro to stimulate the human monocytic leukemia THP-1 cell line or bone marrow-derived macrophages (BMDM) isolated from WT or NLRP3 KO mice. The gene expression of pro- and anti-inflammatory cytokines was evaluated by quantitative PCR; IL-1β, IL-6 and IL-8 production by ELISA; and mitogen-activated protein kinase (MAPK) activation by immunoblot analysis. NF-κB activation was determined in THP-1 cells containing a reporter plasmid. In vivo, the inflammatory potential of CPP phases was assessed with the murine air pouch model via cell analysis and production of IL-1β and CXCL1 in the exudate. The role of NF-κB was determined by a pharmacological approach, both in vivo and in vitro.Results: In vitro, IL-1β production induced by m- and t-CPPD and m-CPPTβ crystals was NLRP3 inflammasome dependent. m-CPPD crystals were the most inflammatory by inducing a faster and higher production and gene expression of IL-1β, IL-6, and IL-8 than t-CPPD, m-CPPTβ and MSU crystals. The a-CPP phase did not show an inflammatory property. Accordingly, m-CPPD crystals led to stronger activation of NF-κB, p38, extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) MAPKs. Inhibition of NF-κB completely abrogated IL-1β and IL-8 synthesis and secretion induced by all CPP crystals. Also, inhibition of JNK and ERK1/2 MAPKs decreased both IL-1β secretion and NF-κB activation induced by CPP crystals. In vivo, IL-1β and CXCL1 production and neutrophil infiltration induced by m-CPPD crystals were greatly decreased by NF-κB inhibitor treatment.Conclusion: Our results suggest that the inflammatory potential of different CPP crystals relies on their ability to activate the MAPK-dependent NF-κB pathway. Studies are ongoing to investigate the underlying mechanisms.

Highlights

  • Calcium pyrophosphate dihydrate (CPPD) microcrystal deposition disease affects more than 17.5% of people over 75 years old [1, 2]

  • On optical microscopy of THP1 cells cultured with CPP phases, m-CPPD crystals exhibited a needle-shaped structure and were the smallest and were more dispersed in cell culture than the others, whereas t-CPPD crystals had a rectangular shape, m-CPPTβ crystals were platelet-like and formed aggregates and the amorphous CPP (a-CPP) phase corresponded to large shapeless particles (Figure 1B)

  • interleukin 1β (IL-1β) and IL-8 production induced by m-CPPD and t-CPPD were both substantially decreased by JNK and extracellular signal-regulated kinase 1/2 (ERK1/2) mitogen-activated protein kinases (MAPKs) inhibitors (Figure 8C) but were not modulated by the p38 MAPK inhibitor (Figure 8D)

Read more

Summary

Introduction

Calcium pyrophosphate dihydrate (CPPD) microcrystal deposition disease affects more than 17.5% of people over 75 years old [1, 2]. Monoclinic and triclinic CPPD phases (m-CPPD and t-CPPD, respectively) are the two types of CPPD crystals identified in synovial fluids at the time of acute arthritis, but the existence of precursor phases such as m-CPP tetrahydrate beta (m-CPPTβ) and amorphous CPP (a-CPP) have been suggested in vitro [3, 4]. Smaller crystals are more active than larger ones: the crystal surface plays a major role in determining cellular response and m-CPPD may be more active than t-CPPD [6,7,8,9,10] In line with these preclinical results, acute inflammatory synovial fluids contain a greater proportion of m-CPPD crystals and low inflammatory samples contain a greater proportion of t-CPPD crystals [7]. Two CPP microcrystals, namely monoclinic and triclinic CPP dihydrates (m- and t-CPPD), have been identified in human tissues in different proportions according to clinical features. m-CPP tetrahydrate beta (m-CPPTβ) and amorphous CPP (a-CPP) phases are considered as m- and t-CPPD crystal precursors in vitro

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