Faculty Opinions recommendation of Complement drives Th17 cell differentiation and triggers autoimmune arthritis.

Author(s):  
Rene Toes ◽  
Leendert Trouw
Oncotarget ◽  
2015 ◽  
Vol 7 (8) ◽  
pp. 8590-8600 ◽  
Author(s):  
Xinyi Tang ◽  
Jie Tian ◽  
Jie Ma ◽  
Jiemin Wang ◽  
Chen Qi ◽  
...  

2017 ◽  
Vol 187 (9) ◽  
pp. 2034-2045 ◽  
Author(s):  
Jin-Sil Park ◽  
Jeong-Hee Jeong ◽  
Jae-Kyeong Byun ◽  
Mi-Ae Lim ◽  
Eun-Kyung Kim ◽  
...  

2019 ◽  
Vol 2019 ◽  
pp. 1-11 ◽  
Author(s):  
Da Som Kim ◽  
Hong-Ki Min ◽  
Eun Kyung Kim ◽  
Seung Cheon Yang ◽  
Hyun Sik Na ◽  
...  

Rheumatoid arthritis (RA) is a type of systemic autoimmune arthritis that causes joint inflammation and destruction. One of the pathological mechanisms of RA is known to involve histone acetylation. Although the histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) can attenuate arthritis in animal models of RA, the mechanism underlying this effect is poorly understood. This study was performed to examine whether SAHA has therapeutic potential in an animal model of RA and to investigate its mechanism of action. Collagen-induced arthritis (CIA) mice were orally administered SAHA daily for 8 weeks and examined for their arthritis score and incidence of arthritis. CD4+ T cell regulation following SAHA treatment was confirmed in splenocytes cultured under type 17 helper T (Th17) cell differentiation conditions. Clinical scores and the incidence of CIA were lower in mice in the SAHA treatment group compared to the controls. In addition, SAHA inhibited Th17 cell differentiation, as well as decreased expression of the Th17 cell-related transcription factors pSTAT3 Y705 and pSTAT3 S727. In vitro experiments showed that SAHA maintained regulatory T (Treg) cells but specifically reduced Th17 cells. The same results were obtained when mouse splenocytes were cultured under Treg cell differentiation conditions and then converted to Th17 cell differentiation conditions. In conclusion, SAHA was confirmed to specifically inhibit Th17 cell differentiation through nuclear receptor subfamily 1 group D member 1 (NR1D1), a factor associated with Th17 differentiation. The results of the present study suggested that SAHA can attenuate CIA development by inhibition of the Th17 population and maintenance of the Treg population through NR1D1 inhibition. Therefore, SAHA is a potential therapeutic candidate for RA.


2010 ◽  
Vol 207 (6) ◽  
pp. 1135-1143 ◽  
Author(s):  
Motomu Hashimoto ◽  
Keiji Hirota ◽  
Hiroyuki Yoshitomi ◽  
Shinji Maeda ◽  
Shin Teradaira ◽  
...  

Activation of serum complement triggers Th17 cell–dependent spontaneous autoimmune disease in an animal model. In genetically autoimmune-prone SKG mice, administration of mannan or β-glucan, both of which activate serum complement, evoked Th17 cell–mediated chronic autoimmune arthritis. C5a, a chief component of complement activation produced via all three complement pathways (i.e., lectin, classical, and alternative), stimulated tissue-resident macrophages, but not dendritic cells, to produce inflammatory cytokines including IL-6, in synergy with Toll-like receptor signaling or, notably, granulocyte/macrophage colony-stimulating factor (GM-CSF). GM-CSF secreted by activated T cells indeed enhanced in vitro IL-6 production by C5a-stimulated macrophages. In vivo, C5a receptor (C5aR) deficiency in SKG mice inhibited the differentiation/expansion of Th17 cells after mannan or β-glucan treatment, and consequently suppressed the development of arthritis. Transfer of SKG T cells induced Th17 cell differentiation/expansion and produced arthritis in C5aR-sufficient recombination activating gene (RAG)−/− mice but not in C5aR-deficient RAG−/− recipients. In vivo macrophage depletion also inhibited disease development in SKG mice. Collectively, the data suggest that complement activation by exogenous or endogenous stimulation can initiate Th17 cell differentiation and expansion in certain autoimmune diseases and presumably in microbial infections. Blockade of C5aR may thus be beneficial for controlling Th17-mediated inflammation and autoimmune disease.


2017 ◽  
Vol 2017 ◽  
pp. 1-13 ◽  
Author(s):  
Jie Yang ◽  
Lidong Liu ◽  
Yiming Yang ◽  
Ning Kong ◽  
Xueyu Jiang ◽  
...  

Objective.Tolerogenic dendritic cells (tDCs) can expand TGF-β-induced regulatory T cells (iTregs); however, the therapeutic utility of these expanded iTregs in autoimmune diseases remains unknown. We sought to determine the properties of iTregs expanded by mature tolerogenic dendritic cells (iTregmtDC) in vitro and explore their potential to ameliorate collagen-induced arthritis (CIA) in a mouse model.Methods. After induction by TGF-βand expansion by mature tDCs (mtDCs), the phenotype and proliferation of iTregmtDCwere assessed by flow cytometry. The ability of iTregs and iTregmtDCto inhibit CD4+T cell proliferation and suppress Th17 cell differentiation was compared. Following adoptive transfer of iTregs and iTregmtDCto mice with CIA, the clinical and histopathologic scores, serum levels of IFN-γ, TNF-α, IL-17, IL-6, IL-10, TGF-βand anti-CII antibodies, and the distribution of the CD4+Th subset were assessed.Results. Compared with iTregs, iTregmtDCexpressed higher levels of Foxp3 and suppressed CD4+T cell proliferation and Th17 cell differentiation to a greater extent. In vivo, iTregmtDCreduced the severity and progression of CIA more significantly than iTregs, which was associated with a modulated inflammatory cytokine profile, reduced anti-CII IgG levels, and polarized Treg/Th17 balance.Conclusion.This study highlights the potential therapeutic utility of iTregmtDCin autoimmune arthritis and should facilitate the future design of iTreg immunotherapeutic strategies.


Sign in / Sign up

Export Citation Format

Share Document