Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2020

Open Access 01.12.2020 | Research

TDAG8 deficiency reduces satellite glial number and pro-inflammatory macrophage number to relieve rheumatoid arthritis disease severity and chronic pain

verfasst von: Shih-Ping Dai, Wei-Shan Hsieh, Chien-Hua Chen, Yueh-Hao Lu, Hsu-Shan Huang, Der-Ming Chang, Shir-Ly Huang, Wei-Hsin Sun

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2020

Abstract

Background

The autoimmune disease rheumatoid arthritis (RA) affects approximately 1% of the global population. RA is characterized with chronic joint inflammation and often associated with chronic pain. The imbalance of pro-inflammatory and anti-inflammatory macrophages is a feature of RA progression. Glial cells affecting neuronal sensitivity at both peripheral and central levels may also be important for RA progression and associated pain. Genetic variants in the T cell death-associated gene 8 (TDAG8) locus are found to associate with spondyloarthritis. TDAG8 was also found involved in RA disease progression and associated hyperalgesia in the RA mouse model. However, its modulation in RA remains unclear.

Methods

To address this question, we intra-articularly injected complete Freund’s adjuvant (CFA) into TDAG8+/+, TDAG8−/− or wild-type mice, followed by pain behavioral tests. Joints and dorsal root ganglia were taken, sectioned, and stained with antibodies to observe the number of immune cells, macrophages, and satellite glial cells (SGCs). For compound treatments, compounds were intraperitoneally or orally administered weekly for 9 consecutive weeks after CFA injection.

Results

We demonstrated that TDAG8 deletion slightly reduced RA pain in the early phase but dramatically attenuated RA progression and pain in the chronic phase (> 7 weeks). TDAG8 deletion inhibited an increase in SGC number and inhibition of SGC function attenuated chronic phase of RA pain, so TDAG8 could regulate SGC number to control chronic pain. TDAG8 deletion also reduced M1 pro-inflammatory macrophage number at 12 weeks, contributing to the attenuation of chronic RA pain. Such results were further confirmed by using salicylanilide derivatives, CCL-2d or LCC-09, to suppress TDAG8 expression and function.

Conclusions

This study demonstrates that TDAG8 deletion reduced SGC and M1 macrophage number to relieve RA disease severity and associated chronic pain. M1 macrophages are critical for the development and maintenance of RA disease and pain, but glial activation is also required for the chronic phase of RA pain.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
RA
Rheumatoid arthritis
TDAG8
T cell death-associated gene 8
CFA
Complete Freund’s adjuvant
SGC
Satellite glial cells
DRG
Dorsal root ganglia
FC
DL-fluorocitric acid
TNF
Tumor necrosis factor
IL-6
Interleukin 6
IL-17
Interleukin 17
Th17
T-helper 17
H&E
Hematoxylin and eosin:
HEK293T
Human embryonic kidney, adenovirus type 5-transformed 293 cells
PERI
Peripherin
GFAP
Glial fibrillary acidic protein
FLS
Fibroblast-like synoviocytes
RANKL
Receptor activator of NF-κB ligand
CGRP
Calcitonin gene-related peptide

Introduction

Rheumatoid arthritis (RA) is an autoimmune disease and characterized with chronic joint inflammation. Chronic joint inflammation causes cartilage damage and ultimately total joint destruction but is also associated with ongoing pain and increased pain [13]. The magnitude of pain may not necessarily be associated with the severity of the underlying disease, and pain may persist even when disease exacerbations have apparently lessened. In RA, the neurophysiological mechanisms underlying pain remain unclear. Experimental animal models of inflammatory arthritis suggest that changes in neuronal sensitivity at both peripheral and central levels may be important [4, 5]. The causes of RA-associated pain could also differ in early and late disease stages. The acute phase of pain could be associated with acute joint inflammation, but the chronic phase could be linked to inflammatory components of neuron–immune interactions and non-inflammatory components such as neuron–glia interactions or central mechanisms [6, 7].
In RA, resident macrophages become activated in synovial tissues; they along with infiltrated macrophages secrete pro-inflammatory cytokines (e.g., tumor necrosis factor α [TNFα], interleukin 6 [IL-6]), mediators, and enzymes to regulate synovial inflammation and joint destruction [8]. Activated macrophages also produce anti-inflammatory cytokines (e.g., IL-10) to promote the resolution of inflammation and tissue repair, thus ameliorating the disease. Two different macrophage phenotypes, M1 (classically activated) and M2 (alternatively activated), are responsible for producing pro- and anti-inflammatory cytokines, respectively [9]. The imbalance between pro- and anti-inflammatory cytokines (M1/M2) could be a key mechanism of rheumatic disease progression. RA patients display a more M1 macrophage profile than do people with spondyloarthritis such as psoriatic arthritis [10] or osteoarthritis [11]. An acute hypoxia environment favors M2 macrophage polarization, but chronic hypoxia triggers M1 polarization [12].
In a monoarthritis or collagenase arthritis model, arthritic rats showed satellite glial cell (SGC) proliferation and activation in dorsal root ganglia (DRG) [13, 14]. Intrathecal injection of a glial inhibitor (fluorocitrate) blocked collagenase-induced nociception [14], which suggests that SGCs play some roles in arthritic pain. IL-17A may act on glial cells to sensitize neuron function [15]. Genome-wide association studies demonstrated that genetic variants in the T cell death-associated gene 8 (TDAG8) locus are associated with spondyloarthritis [16], and T-helper 17 (Th17) cells in spondyloarthritis patients show high expression of TDAG8 gene [17]. TDAG8 gene-deficient mice show reduced number of Th17 cells and secretion of IL-17A [18]. Consistent with these data, RA disease severity and RA-evoked pain were attenuated in TDAG8 gene-deficient mice in the RA mouse model [19]. However, how TDAG8 regulates RA and RA-evoked pain in the early and late pain phases remains unclear.
In this study, we used a previously established arthritis mouse model [19] in TDAG8 gene-deficient mice to investigate whether TDAG8-modulated chronic pain and disease severity is related to immune cells or glial cells. TDAG8 gene deficiency reduced SGC number, and SGC inhibition attenuated the chronic phase of RA pain, which suggests that TDAG8 deficiency relieved the late phase of RA pain by regulating SGCs in part. Moreover, reduced M1 macrophage number but not synovial macrophage number in TDAG8–deficient mice may explain the less attenuation of acute-phase RA pain. Consistent with the results of TDAG8 deletion, long-term suppression of TDAG8 gene expression and function by using previously developed salicylanilide derivatives [20, 21], CCL-2d, and LCC-09, reduced RA pain by modulating the number of SGCs and pro-inflammatory macrophages. Accordingly, TDAG8 gene deficiency relieved RA disease severity and pain by reducing SGC number and pro-inflammatory macrophage number.

Materials and methods

Agents

Complete Freund’s adjuvant (CFA) and DL-fluorocitric acid (FC) barium salt were from Sigma-Aldrich. Salicylanilide derivatives CCL-2d (3-(4-Chloro-2-fluorophenyl)-7-methoxy-2H-benzo[e][1,3]-oxazine-2,4(3H)-dione) and LCC-09 (N-(3-cyanophenyl)-20,40-difluoro-4-hydroxy-[1,10-biphenyl]-3-carboxamide) were synthesized as described [20, 21]. Tofacitinib (commercial RA drug) [22] was from Selleckchem. For animal experiments, all drugs or compounds were first solved in dimethylsulfoxide and then diluted in saline before injection.

Arthritis mouse model

Male or female ICR mice (8–12 weeks old) were purchased from BioLASCO Taiwan (Taipei) and housed 3–4 per cage under a 12-h light/dark cycle (lights on at 7:00 am) with food and water ad libitum in a temperature- and humidity-controlled environment at National Central University. TDAG8−/− and TDAG8+/+ mice on a B6 background were generated as described [23]. The genotyping primer sequences for TDAG8−/− were 5′-gaaccattagtttggctcatgtgactg/5′-cttgtgtcatgcacaaagtagatgtcc and for TDAG8+/+, 5′-cgaactctagctggcttttatccaataat/5′-gaaccattagtttggctcatgtgactg. Care and use of mice conformed to the Guide for the Use of Laboratory Animals (US National Research Council) and the experimental procedures were approved by the local animal use committee (IACUC, National Central University, Taiwan).
All behavioral testing was performed between 9:00 am and 5:00 pm. Efforts were made to minimize the number of animals used and their suffering. Arthritis was induced as described [19]. Briefly, TDAG8+/+, TDAG8−/− or wild-type (WT ICR) mice were injected with 5 μg CFA in the right ankle joint once a week for 4 weeks (CFA-ctrl). Compounds (CCL-2d [360, 3600 μg/kg], LCC-09 [39, 390 μg/kg], was intraperitoneally injected once at 4w after CFA injection, followed by mechanical tests. For long-term treatment, compounds (CCL-2d [360, 3600 μg/kg], LCC-09 [39, 390 μg/kg], 3 mg/kg tofacitinib) or saline (vehicle) was intraperitoneally or orally (using oral feeding needle, ST-F173 ψ0.9 mm × L 70 mm) administered weekly for 9 consecutive weeks after CFA injection. Some experiments were only intraperitoneally injected once with CCL-2d and LCC-09, followed by the rota-rod tests (by the Taiwan Mouse clinic, Taiwan). FC (0.01 mM) was intrathecally administered at week 3 after CFA injection. Behavioral tests for mechanical or thermal stimuli were performed before and after CFA injection. In some experiments, L4-6 DRG were excised for measuring gene expression, and joints were fixed for hematoxylin and eosin (H&E) staining or immunostaining.

Immunohistochemistry and immunostaining

The severity of the arthritis was scored from 0 to 5 as previously described [19]. Each limb was graded and given a maximum possible score of 15; the maximum score for an animal was 60.
Histological staining was performed as previously described [19]. Briefly, at 12 weeks after CFA injection, the tibiotarsal joint was excised, fixed, decalcified, embedded in paraffin and sectioned longitudinally at 10 μm with use of a microtome, then stained with HE (by the Taiwan Mouse Clinic, Taipei). Images were observed by light microscopy (Lecia, LAS EZ). Arthritic changes were scored on a scale of 0 to 5 as previously described [19]. From each joint, 6 areas of 2 sections were used to provide a representative sample of the whole joint. Cell number counted is dependent on different type of macrophages, so cell density (cells/mm2) is used to present the data. Mean scores were the average of all section scores for each animal. Three animals are used for each point.
Some joint sections were stained with the antibodies for CD80 (1:250, Biorbyt, UK), CD68 (1:100, Biorbyt, UK) or CD163 (1:100, Biorbyt, UK), followed by alkaline phosphatase-conjugated goat anti-rabbit IgG (1:5000, Jackson Immunoresearch). Signals were developed by nitro-blue tetrazolium chloride and 5-bromo-4-chloro-3′-Indolyphosphate p-toluidine (Millipore). Immunoreactivity-positive cells were counted.
Staining of SGCs was as previously described [24]. Briefly, at 0, 1, 4, 8, and 12 weeks after CFA injection, DRG isolated from vehicle- or compound-treated mice were frozen in freezing solution and cut at 12 μm by using a cryostat (Leica microsystem 3510S, Bensheim, Germany). Sections were co-stained with the antibodies for peripherin (PERI, 1:500, Sigma) and glial fibrillary acidic protein (GFAP; 1:1000; Dako), followed by TRITC-conjugated goat–anti-mouse IgG antibody (1:250, Sigma) and FITC-conjugated goat-anti-rabbit-IgG antibody (1:250, Sigma), respectively. The digitized images were captured by using MetaVue. PERI-positive neurons surrounded by GFAP-positive SGCs in one-third or more of the PERI circumference were counted (PERIGFAP+) and expressed as a percentage of total PERI-IR neurons (PERIT) in the fields analyzed (PERIGFAP+/PERIT). Data for each treatment group were collected from 10 DRG sections. The distance between two sections at least 60 μm. Total 1000–2000 PERI-positive neurons were counted for each group.

Assessment of arthritic pain in mice

Pain behavioural tests were as described previously [25]. Briefly, mice were tested for withdrawal thresholds to mechanical stimuli applied to the plantar aspect of the hindpaw. Mice were pre-trained for 2 h each day and for 3 days before the test. Before and after CFA injection, a series (ascending force) of von Frey fibers (Touch-Test, North Coast Medical, Morgan Hill, CA) was applied. A von Frey fiber was applied to each paw 5 times at 5-s intervals. The paw withdrawal threshold (PWT) was when paw withdrawal was observed in more than 3 of 5 applications.
For thermal nociceptive response to radiant heat applied to the plantar surface of the paw, before and after CFA injection, the plantar surface of mouse hindpaws was stimulated with a lit light bulb (30% intensity, 25 s for cut-off time). The latency to withdrawal of the paw (PWL) from radiant heat was measured. Measurements from three trials at 1-min intervals in each paw were averaged. The mean basal withdrawal latencies of 15~20 s was obtained in non-injected mice.

Measurement of cytokine levels in serum

Mice with or without compound treatments were sacrificed at 8 or 12 weeks. Blood was collected by cardiac puncture. For serum samples, blood was left to clot for 30min at 4 °C, followed by centrifugation for 20 min at 2000×g. Serum was aliquoted and stored at – 80 °C. TNF-α or IL-6 levels were measured with kits from R&D systems (Minneapolis, MN, USA).

Quantitative RT-PCR

Lumbar 1–5 (L1–5) DRG ipsilateral and contralateral to injected paws were removed at 0 or 12 weeks for RNA extraction, with DRG from 0 weeks as a control. RNA extraction was performed as described [26]. Each DRG pool of each point contained at least 10 DRG from one side of 3 mice. RNA was extracted by using the RNeasy kit (Qiagen, Valencia, CA, USA). Each gene primer (100 nM), derived cDNA, and master mix (SYBR green I and AmpliTaq Gold DNA polymerase [Applied Biosystems, Foster City, CA, USA]) were mixed for PCR reactions and product detection by using the ABI Prism 7300 system. For each assay, preparations were run in triplicate. The thermal cycling conditions were 95 °C for 10 min, followed by 40 cycles of 95 °C for 15 s, and 60 °C for 1 min. The threshold cycle (Ct) values for both targets and the internal reference (mGAPDH) were measured from the same samples, and the expression of target genes relative to that of mGAPDH was calculated by the comparative Ct method.
The primer sequences for TDAG8 (197 bp) were 5′-atagtcagcgtcccagccaac (forward)/5′-cgcttcctttgcacaaggtg (reverse) and for mGAPDH (233 bp), 5′-ggagccaaacgggtcatcatctc (forward)/ 5′-gaggggccatccacagtcttct (reverse).

Calcium imaging

To detect TDAG8-mediated signaling, calcium imaging was performed as described [24]. Briefly, human embryonic kidney, adenovirus type 5-transformed 293 cells (HEK293T, obtained from the Bioresource Collection and Research Center of Food Industry Research and Development Institute, Taiwan) were cultured on coverslips and transfected with 1.2 μg pIRES-GFP-TDAG8. Transfected cells were pre-incubated with 2.5 μM Fura-2 acetoxymethyl ester (Fura-2-AM, Molecular Probes) for 40 min in HEPES/MES buffer (125 mM NaCl, 1 mM KCl, 5 mM CaCl2, 1 mM MgCl2, 8 mM glucose, 10 mM HEPES, and 15 mM MES, pH7.6). After washing, cells were stimulated with HEPES/MES (pH 5.5) buffer, followed by [Ca2+]i recording with a Leica DMI3000B fluorescence microscope and a Ca2+ imaging system and analyzed by using MetaFluor software. The fluorescence ratio at two excitation wavelengths (340/380 nm, Ca2+-bound Fura-2-AM/free Fura-2-AM) was recorded and analyzed. The pH-evoked calcium transients and number of cells responding to the indicated pH values were recorded. For compound treatments, cells were stimulated with HEPES/MES buffer (pH 5.5) containing different concentrations of compounds.

Statistical analysis

All data are presented as mean ± SEM. One-way or two-way ANOVA with post hoc Bonferroni correction was used to compare results from multiple groups. Non-parametric Mann-Whitney U test was used to compare results from two groups. For SGC number analysis, the z test for two proportions was used to test the level of significance, with 95% confidence intervals estimated. p < 0.05 was considered statistically significant. A statistical power analysis was performed for sample size estimation, based on data from pilot study, comparing TDAG8−/− to TDAG8+/+ group. The effect size (ES) for t test was 3.4 and for two-way ANOVA was 0.30. With an alpha = 0.05 and power = 0.80, the sample size needed with the effect size (GPower 3.1) was approximately N = 3/group for the t test or total N = 48 for two–way ANOVA. Thus, our sample size of N ≥ 3 or N ≥ 48 was adequate.

Results

RA mice display increased number of SGCs

RA mice were established as previously described [19] and exhibited long-term bilateral mechanical (Fig. 1a) and thermal hyperalgesia (Fig. 1b). In the first week after CFA injection, the mean arthritis score (measuring joint, toe, and paw swelling of four limbs) was 16.1 ± 2.9% (maximum 100%). The severity of joint swelling increased with time and reached a peak at 4 weeks (42.8 ± 1.7%), remained for 8 weeks and slightly decreased at 12 weeks (25.8 ± 1.3%) (Fig. 1c). In our previous study, no synovial inflammation and swelling in saline-injected joints [19]. In RA mice, we observed swelling in uninjected joints as arthritis scores are more than one limb (each limb for 25%, arthritis scores are 42% after 4 weeks), although we did not detect dramatic synovial inflammation in uninjected joints. However, we did find bilateral hyperalgesia and raised serum cytokines TNFα, IL-7, and IL6 in arthritis mice. It suggests that polyarthritis occurred in RA mice. In a monoarthritis model or collagenase arthritis model, arthritic rats showed SGC proliferation and activation [13, 14]. To determine whether the number of SGCs was also increased in our RA mice, DRG from RA mice were immunostained for anti-GFAP (a SGC marker). Most GFAP(+) cells surrounded PERI(+) (a nociceptor marker) neurons (Fig. 1d). The number of PERI(+) neurons surrounded by GFAP(+) cells (PERIGFAP+) was increased and peaked in the first week, then declined slightly later (Fig. 1e).

TDAG8 deletion reduces mechanical and thermal hyperalgesia and arthritis scores

We previously found that TDAG8 is involved in RA-induced pain and disease progression [19]. Here we used TDAG8 gene-deficient mice to understand how TDAG8 regulates RA. TDAG8 gene deficiency attenuated bilateral mechanical hyperalgesia from week 7 on the ipsilateral side (Fig. 2a) and from week 8 on the contralateral side (Fig. 2b). Bilateral thermal hyperalgesia was attenuated from week 6 at both sides (Fig. 2c, d). Arthritis scores were reduced from week 4 (Fig. 2e). IL-6 production was inhibited at 12 weeks (Fig. 2f) and IL-17 production at 8 weeks in TDAG8-deficient mice (Fig. 2g).
Consistent with reduced arthritic scores in TDAG8−/− mice, except for pannus, bone destruction and cartilage damage was greatly attenuated in TDAG8−/− mice at week 12 (Fig. 3A–D). We used three macrophage markers to examine macrophage infiltration in joints: CD68+ for synovial macrophages and intimal fibroblast-like synoviocytes (FLSs), CD80+ for pro-inflammatory M1 macrophages, and CD163+ for anti-inflammatory M2 macrophages. We previously found increased CD68+ and CD80+ cell number with time, corresponding to RA disease progression in mice [19]. Here, TDAG8−/− mice did not show reduced CD68+ cell number (Fig. 3E, H), which may reflect lack of significantly reduced pannus. M1 (CD80+) macrophage number was significantly decreased at week 12 (Fig. 3E, F), but M2 (CD163+) macrophage number remained unchanged (Fig. 3E, G).
Similar to wild-type RA mice, TDAG8+/+ RA mice showed increased number of SGCs, which peaked at the first week (Fig. 4a, b). Nevertheless, the number of SGCs was not increased over time in TDAG8−/− mice (Fig. 4a, b). Intrathecal administration of the SGC inhibitor FC at week 3 after CFA injection attenuated mechanical hyperalgesia from week 10 (Fig. 4c), which suggests that SGCs contribute to the chronic phase of RA-evoked hyperalgesia.

CCL-2d and LCC-09 relieve RA-induced hyperalgesia

To further confirm the TDAG8 involvement in RA disease and pain, we tested the previously developed small salicylanilide derivatives CCL-2d (Fig. 5a) and LCC-09 (Fig. 6a) [20, 21] in RA mice. After CFA injection for 4 weeks, mice were intraperitoneally (i.p.) injected once with the compounds at different doses, then underwent a rota-rod test or a mechanical behavioral test. The dosages of CCL-2d were determined by in vitro cell-based and in vivo animal experiments (data not shown). The rotarod results showed no differences between vehicle-treated and CCL-2d-treated mice (Fig. 5b). One-time administration of CCL2-d reduced mechanical hyperalgesia from 30 min, peaked at 60 min and declined at 180 min (Fig. 5c). Doses of 360 and 3600 μg/kg CCL-2d attenuated mechanical hyperalgesia at 60 min (Fig. 5d). CCL-2d 360 μg/kg injected weekly for 9 consecutive weeks effectively reduced the mechanical hyperalgesia in both ipsilateral and contralateral sides, and the analgesic effects improved up to week 9 (Fig. 5e, f), then decreased. The CCL-2d–induced analgesic effect seemed cumulative because the paw withdrawal threshold (PWT) before CCL-2d injection (B) was increased from 6 weeks. As compared with tofacitinib (commercial RA drug, 3 mg/kg), CCL-2d (360 μg/kg) had a greater analgesic effect from weeks 5 to 9 on the ipsilateral side (Fig. 5g, h). Oral administration of CCL-2d also attenuated the mechanical hyperalgesia (Fig. 5i, j), and the analgesic effect of oral administration was similar to that of tofacitinib.
Similar to CCL-2d, no difference between vehicle-treated and LCC-09-treated mice was found in rota-rod tests (Fig. 6b). One-time administration of LCC-09 (390 μg/kg) reduced mechanical hyperalgesia from 30 min, peaked at 60 min and then declined (Fig. 6c). Injection of LCC-09 at 39 or 390 μg/kg reduced mechanical hyperalgesia at 60 min (Fig. 6d). LCC-09 390 μg/kg injected weekly for 9 consecutive weeks effectively attenuated the mechanical hyperalgesia on both sides and the analgesic effects improved up to week 12 (Fig. 6e, f). The analgesic effect of LCC-09 was similar to that of tofacitinib (3 mg/kg) (Fig. 6g, h). Similar results were found with oral administration of LCC-09 (Fig. 6i, j).

CCL-2d and LCC-09 reduce synovial inflammation, bone erosion, and macrophage infiltration

Histopathological examination of the ankle joints of CCL-2d–, LCC-09–, or tofacitinib-treated mice at 12 weeks revealed that CCL-2d or LCC-09 but not tofacitinib reduced pannus and bone erosion induced by repeated CFA injection (Fig. 7A–D). Pannus was significantly reduced with LCC-09 (Fig. 7B), and bone erosion was significantly decreased with CCL-2d (Fig. 7C). CCL-2d treatment also decreased serum IL-6 level at 12 weeks (Fig. 7E), and both treatments reduced serum TNF-α level at 12 weeks (Fig. 7F). Tofacitinib did not reduce the serum level of IL-6 or TNF-α (Fig. 7E, F).
CCL-2d and LCC-09 treatment had distinct effects on macrophages and FLSs. Only LCC-09 significantly reduced CD68+ cell number (Fig. 8a, b), which was consistent with reduced pannus (Fig. 8b). However, both LCC-09 and CCL-2d decreased CD80+ cell number, and CCL-2d increased CD163+ cell number (Fig. 8a, c, d). CD163+ macrophages have an anti-inflammatory role, which could explain why CCL-2d treatment did not significantly reduce pannus but decreased bone-erosion and serum IL-6 levels. Although tofacitinib reduced CD80+ cell number 33%, it did not reduce the CD68+ cell number, which could explain the lack of reduction in pannus with tofacitinib (Fig. 8a–c).

CCL-2d and LCC-09 suppress TDAG8 gene expression and SGC number

To understand the mechanism of the analgesic action of CCL-2d or LCC-09, we used DRG from RA mice with or without CCL-2d or LCC-09 treatment and examined TDAG8 gene expression. TDAG8 mRNA expression was increased at 12 weeks in RA mice, which is consistent with our previous study [19]. Both CCL-2d and LCC-09 reduced this increase in expression to the basal level (Fig. 9a). Because both compounds inhibited TDAG8 gene expression, we then examined whether CCL-2d or LCC-09 could inhibit TDAG8 function. Since TDAG8 activation induces cAMP accumulation and intracellular calcium increase [23], we examined TDAG8 function using calcium imaging to detect intracellular calcium increase. TDAG8 was transfected into HEK293 cells. Transfected cells were treated with 1, 10, and 100 nM CCL-2d or LCC-09. CCL-2d 10 nM partially inhibited acid-induced signals in TDAG8-expressing cells, but 100 nM completely inhibited TDAG8-mediated signaling (Fig. 9b). LCC-09 100 nM also inhibited TDAG8-mediated signaling. Hence, CCL-2d and LCC-09 may both inhibit TDAG8 gene expression and function.
We then examined whether CCL-2d or LCC-09 could suppress TDAG8 expression and function to control SGC number. CCL-2d and LCC-09 treatment attenuated the increased number of SGCs at week 12 (Fig. 9c–f), which suggests that CCL2d and LCC-09 could act on TDAG8 to inhibit SGC activation, further regulating bilateral hyperalgesia.

Discussion

We previously found TDAG8 involved in RA disease progression and associated pain [19]. Here, we demonstrated that RA development increased the number of SGCs, and inhibition of glial activation blocked the chronic phase of RA-associated pain. TDAG8 gene deletion inhibited SGC activation and attenuated the chronic phase of RA-associated pain. Thus, TDAG8 may modulate activation of SGCs to attenuate the chronic phase of RA-associated pain. In addition, TDAG8 deletion reduced the number of pro-inflammatory M1 macrophages, which may explain the decrease in arthritis scores and associated pain in TDAG8-deficient mice. We examined the effects of two salicylanilide derivatives, CCL-2d and LCC-09, which were originally designed to inhibit receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis [20, 21] in RA disease progression and associated pain. Both compounds attenuated RA progression and associated pain. These two compounds inhibited TDAG8 gene expression in RA mice and blocked TDAG8 signalling, thereby reducing the number of SGCs and pro-inflammatory M1 macrophages. Thus, TDAG8 may control the number of SGCs and proinflammatory macrophages to modulate RA disease severity and associated pain.
TDAG8 gene deletion greatly attenuated chronic hyperalgesia (> 7 weeks) but less effectively acute hyperalgesia in our mouse model. TDAG8 may regulate different components contributing to the early and late RA pain stages. The acute phase of pain could be largely attributed to acute joint inflammation because of an increase in synovial macrophage number. TDAG8 deficiency reduced the number of pro-inflammatory M1 macrophages (CD80+ cells) but not synovial macrophages (CD68+ cells). Therefore, the acute hyperalgesia was less attenuated in TDAG8−/− mice, which may explain why arthritis scores were not reduced in the early stage of RA in TDAG8−/− mice. The hypoxia condition may favor M1 polarization [12]. Given that TDAG8 is expressed in macrophages [2729] and TDAG8 responds to acid [30, 31], mice lacking the TDAG8 gene may have reduced macrophage responsiveness to the hypoxia condition, which would not favor M1 polarization.
In the chronic phase, RA progression mainly depends on the increase in M1 macrophage number and cytokine levels (IL-17 and IL-6). Given that both IL-6 and IL-17 are involved in arthritis-induced hyperalgesia [3234] and administration of a macrophage blocker reduced hyperalgesia [35], the reduced M1 macrophage number and IL-6 and IL-17 levels we showed may have synergistically contributed to a decline in chronic hyperalgesia and arthritis score. Moreover, neuron–glia interactions play a role in the chronic phase. Consistent with previous studies in monoarthritis or osteoarthritis [13, 14], our RA mice showed increased number of SGCs. With TDAG8 deficiency, the increased number of SGCs was completely inhibited. Administration of an SGC inhibitor at week 3 attenuated the chronic hyperalgesia (from 6 weeks). Reduced chronic hyperalgesia caused by TDAG8 deletion could also be attributed to an inhibition of the increased SGC number. This suggestion could explain why hyperalgesia was greatly attenuated in the late phase in TDAG8−/− mice. SGCs can be activated by substance P or calcitonin gene-related peptide (CGRP) [36] or by IL-17 [15]. Given that TDAG8 gene knockdown in a peripheral nerve reduces neuron activity [23], TDAG8 deficiency may reduce neuron activity and release of substance P or CGRP, thereby blocking SGC activation. TDAG8 gene-deficient mice show reduced number of Th17 cells and secretion of IL-17A [18]. Alternatively, TDAG8 deficiency could decrease IL-17 levels to directly attenuate RA-induced hyperalgesia or indirectly affect SGC activation thereby reducing hyperalgesia. TDAG8−/− mice showed reduced IL-17 level from week 8, which corresponded to attenuated hyperalgesia in the chronic phase.
A previous study of TDAG8–deficient mice found that TDAG8 deficiency promotes arthritis using anti-collagen antibody/lipopolysaccharide model [37], suggesting that TDAG8 is a negative regulator for RA. However, recent studies in human patients found that spondyloarthritis patients are associated with genetic variants of TDAG8 locus [16], and their Th17 cells show high expression of TDAG8 gene [17]. TDAG8 gene-deficient mice show reduced number of Th17 cells and secretion of IL-17A [18]. These results suggest that TDAG8 is a positive regulator for RA progression, which are consistent with our RA animal results.
Similar to results from TDAG8-deficient mice, consecutive administration of the salicylanilide derivative CCL-2d or LCC-09 suppressed TDAG8 expression and function, further reducing the bilateral mechanical hyperalgesia induced by arthritis. Both CCL-2d and LCC-09 treatments also reduced the number of SGCs and M1 macrophages. These two compounds likely act on TDAG8 to reduce the proliferation of SGCs and M1 macrophage polarization, thereby affecting bilateral hyperalgesia.
Low doses of CCL-2d (360 μg/kg) or LCC-09 (390 μg/kg) had similar analgesic effects as 3 mg/kg tofacitinib injection, which suggests that CCL-2d or LCC-09 could be more effective analgesics than tofacitinib. The analgesic effect of CCL-2d or LCC-09 was transient (lasted 90–120 min) for a single-dose injection, but consecutive injection (once per week) seemed to have synergistic effects in later weeks. Mechanical hyperalgesia before each injection (CCL-2d 0 min or LCC-09 0 min) was gradually attenuated after week 5 (after the first injection). The reason for the synergistic effects is unclear. Given that both compounds inhibit TDAG8 expression and TDAG8-deficient mice also showed gradually attenuated mechanical hyperalgesia from week 7, the synergistic effects of CCL-2d or LCC-09 on hyperalgesia could be attributed to long-term suppression of TDAG8 gene expression and function.
Although TDAG8 gene deletion significantly reduced RA pain, arthritis scores, bone erosion and cartilage damage, it only slightly reduced pannus. Less reduction in synovial inflammation could be due to distinct effects on macrophages and synovial fibroblasts that are predominantly expressed in the synovial lining of the joint and play many roles in synovial inflammation [38, 39]. Both synovial macrophages and intimal fibroblast-like synoviocytes are positive for CD68, whereas CD80 recognizes macrophages in synovial sublinings, bone, and other joint areas. Given that TDAG8 deficiency reduced the increased number of CD80+ macrophages with no inhibition of CD68+ macrophages, TDAG8 may regulate macrophages but not synoviocytes, which explains the slight reduction of pannus. Similarly, CCL-2d greatly decreased CD80+ but not CD68+ cell number, for only a slight reduction in pannus. In contrast, LCC-09 significantly decreased CD68+ and CD80+ cell number, which agrees with the reduced pannus.
The presence of osteoclasts, a form of macrophages in bone, seems essential for RA-induced bone erosion because articular bone remains well preserved in the absence of osteoclasts despite overexpression of proinflammatory cytokines [40]. TDAG8 deficiency reduced CD80+ cell number, so it probably also decreased osteoclasts to prevent bone erosion. Acidosis in joints induces chondrocyte apoptosis [4143], which may explain the prevention of cartilage damage in TDAG8-deficient mice.
In our previous study of TDAG8, gene expression suppressed in peripheral nerves of mice, only the initial phase of RA-induced pain was reduced with TDAG8 knockdown [19]. Nervertheless, in the current study, both the initial and chronic phases of RA pain were reduced in TDAG8-deficient mice. Given that suppression of TDAG8 reduced CD68+ and CD80+ cell number [19] but TDAG8 gene deficiency decreased only CD80+ cell number, CD68+ cells could be the major factor for the initial development of RA pain rather than the chronic phase, whereas CD80+ could contribute to both the initial and chronic phase of RA pain. The finding also explains the attenuated initial arthritis scores in TDAG8-knockdown mice but not TDAG8 gene-deficient mice. Alternatively, we have previously found that ASIC3, TRPV1, and TDAG8 are involved in RA progression and associated pain [19]. Therefore, we have examined ASIC3 and TRPV1 gene expression in TDAG8 knockout mice without RA induction and found no significant change in ASIC3 and TRPV1 gene expression. It suggests that no ASIC3 and TRPV1 influence on TDAG8 knockout mice before RA induction, although it cannot be excluded a possibility that TDAG8 knockout mice have declined expression levels of ASIC3 and TRPV1 genes after RA induction. In both ASIC3 and TRPV1 knockout mice with attenuated chronic hyperalgesia, expression levels of all three genes were eliminated or at low levels at 12 weeks after RA induction. It is likely that high expression levels of ASIC3, TRPV1, and TDAG8 at 12 weeks after RA induction are essential for maintenance of chronic hyperalgesia.
Accordingly, TDAG8 gene expression in peripheral nerves could be critical to regulate CD68+ and CD80+ cell number, thus contributing to the initial development of RA and associated pain, and TDAG8-mediated satellite glial activation and increased IL-6 and IL-17 levels could be essential for maintenance of RA pain.

Conclusions

This study demonstrated that TDAG8 gene deficiency reduced SGC number, and SGC inhibition attenuated the chronic phase of RA pain, which suggests that TDAG8 deficiency relieved the late phase of RA pain by regulating SGCs in part. Moreover, reduced M1 macrophage number but not synovial macrophage number in TDAG8-deficient mice may explain the less attenuation of the acute phase of RA disease activity and associated pain. These results reveal how the acute and chronic phases of RA pain are regulated by TDAG8 through M1 macrophages and SGCs. M1 macrophages are critical for the development and maintenance of RA disease and pain, but statellite glial activation is also required for the chronic phase of RA pain. These findings provide further insights for developing threapeutic treatments for RA disease and associated pain at different stages.

Acknowledgements

We thank the Taiwan Mouse Clinic (MOST 106-2319-B-001-004; MOST 107-2319-B-001-002), which is funded by the National Research Program for Biopharmaceuticals (NRPB) at the Ministry of Science and Technology (MOST) of Taiwan, for technical support in H&E staining.
Care and use of mice conformed to the Guide for the Use of Laboratory Animals (US National Research Council) and the experimental procedures were approved by the local animal use committee (IACUC, National Central University, Taiwan, no. NCU-105-008; NCU-107-002).
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Morris VH, Cruwys SC, Kidd BL. Characterization of capsacin-induced mechanical hyperlagesia as a marker for altered nociceptive processing in patients with rheumatoid arthritis. Pain. 1997;71:179–86.PubMedCrossRef Morris VH, Cruwys SC, Kidd BL. Characterization of capsacin-induced mechanical hyperlagesia as a marker for altered nociceptive processing in patients with rheumatoid arthritis. Pain. 1997;71:179–86.PubMedCrossRef
4.
5.
Zurück zum Zitat Ebersberger A, Grubb BD, Willingale HL, Gardiner NJ, Nebe J, Schaible HG. The intraspinal release of prostaglandin E2 in a model of acute arthritis is accompanied by an up-regulation of cyclo-oxygenase-2 in the spinal cord. Neuroscience. 1999;93(2):775–81.PubMedCrossRef Ebersberger A, Grubb BD, Willingale HL, Gardiner NJ, Nebe J, Schaible HG. The intraspinal release of prostaglandin E2 in a model of acute arthritis is accompanied by an up-regulation of cyclo-oxygenase-2 in the spinal cord. Neuroscience. 1999;93(2):775–81.PubMedCrossRef
7.
Zurück zum Zitat Pinho-Ribeiro FA, Verri WA Jr, Chiu IM. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol. 2017;38(1):5–19.PubMedCrossRef Pinho-Ribeiro FA, Verri WA Jr, Chiu IM. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol. 2017;38(1):5–19.PubMedCrossRef
9.
Zurück zum Zitat Hamilton JA, Tak PP. Analysis of the synovial cell infiltrate in early rheumatoid synovial tissue in relation to local disease activity. Arthritis Rheum. 2009;60:1210–21.PubMedCrossRef Hamilton JA, Tak PP. Analysis of the synovial cell infiltrate in early rheumatoid synovial tissue in relation to local disease activity. Arthritis Rheum. 2009;60:1210–21.PubMedCrossRef
10.
Zurück zum Zitat Vandooren B, Noordenbos T, Ambarus C, Krausz S, Cantaert T, Yeremenko N, Boumans M, Lutter R, Tak PP, Baeten D. Absence of a classically activated macrophage cytokine signature in peripheral spondylarthritis, including psoriatic arthritis. Arthritis Rheum. 2009;60:966–75.PubMedCrossRef Vandooren B, Noordenbos T, Ambarus C, Krausz S, Cantaert T, Yeremenko N, Boumans M, Lutter R, Tak PP, Baeten D. Absence of a classically activated macrophage cytokine signature in peripheral spondylarthritis, including psoriatic arthritis. Arthritis Rheum. 2009;60:966–75.PubMedCrossRef
11.
Zurück zum Zitat Tsuneyoshi Y, Tanaka M, Nagai T, Sunahara N, Matsuda T, Sonoda T, Ijiri K, Komiya S, Matsuyama T. Functional folate receptor beta-expressing macrophages in osteoarthritis synovium and their M1/M2 expression profiles. Scand J Rheumatol. 2012;41:132–40.PubMedCrossRef Tsuneyoshi Y, Tanaka M, Nagai T, Sunahara N, Matsuda T, Sonoda T, Ijiri K, Komiya S, Matsuyama T. Functional folate receptor beta-expressing macrophages in osteoarthritis synovium and their M1/M2 expression profiles. Scand J Rheumatol. 2012;41:132–40.PubMedCrossRef
12.
Zurück zum Zitat Raggi F, Pelassa S, Pierobon D, Penco F, Gattorno M, Novelli F, Eva A, Varesio L, Giovarelli M, Bosco MC. Regulation of Human Macrophage M1-M2 Polarization Balance by Hypoxia and the Triggering Receptor Expressed on Myeloid Cells-1. Front Immunol. 2017;8:1097.PubMedPubMedCentralCrossRef Raggi F, Pelassa S, Pierobon D, Penco F, Gattorno M, Novelli F, Eva A, Varesio L, Giovarelli M, Bosco MC. Regulation of Human Macrophage M1-M2 Polarization Balance by Hypoxia and the Triggering Receptor Expressed on Myeloid Cells-1. Front Immunol. 2017;8:1097.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Nascimento DS, Castro-Lopes JM, Moreira Neto FL. Satellite glial cells surrounding primary afferent neurons are activated and proliferate during monoarthritis in rats: is there a role for ATF3? PLoS One. 2014;9:e108152.PubMedPubMedCentralCrossRef Nascimento DS, Castro-Lopes JM, Moreira Neto FL. Satellite glial cells surrounding primary afferent neurons are activated and proliferate during monoarthritis in rats: is there a role for ATF3? PLoS One. 2014;9:e108152.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Adães S, Almeida L, Potes CS, Ferreira AR, Castro-Lopes JM, Ferreira-Gomes J, Neto FL. Glial activation in the collagenase model of nociception associated with osteoarthritis. Mol Pain. 2017;13:1–12.CrossRef Adães S, Almeida L, Potes CS, Ferreira AR, Castro-Lopes JM, Ferreira-Gomes J, Neto FL. Glial activation in the collagenase model of nociception associated with osteoarthritis. Mol Pain. 2017;13:1–12.CrossRef
15.
Zurück zum Zitat Moynes DM, Vanner SJ, Lomax AE. Participation of interleukin 17A in neuroimmune interactions. Brain Behav Immun. 2014;41:1–9.PubMedCrossRef Moynes DM, Vanner SJ, Lomax AE. Participation of interleukin 17A in neuroimmune interactions. Brain Behav Immun. 2014;41:1–9.PubMedCrossRef
16.
Zurück zum Zitat Cortes A, Hadler J, Pointon JP, Robinson PC, Karaderi T, Leo P, Cremin K, Pryce K, Harris J, Lee S, Joo KB, Shim SC, Weisman M, Ward M, Zhou X, Garchon HJ, Chiocchia G, Nossent J, Lie BA, Førre Ø, Tuomilehto J, Laiho K, Jiang L, Liu Y, Wu X, Bradbury LA, Elewaut D, Burgos-Vargas R, Stebbings S, Appleton L, Farrah C, Lau J, Kenna TJ, Haroon N, Ferreira MA, Yang J, Mulero J, Fernandez-Sueiro JL, Gonzalez-Gay MA, Lopez-Larrea C, Deloukas P, Donnelly P, Australo-Anglo-American Spondyloarthritis Consortium (TASC); Groupe Française d'Etude Génétique des Spondylarthrites (GFEGS); Nord-Trøndelag Health Study (HUNT); Spondyloarthritis Research Consortium of Canada (SPARCC); Wellcome Trust Case Control Consortium 2 (WTCCC2), Bowness P, Gafney K, Gaston H, Gladman DD, Rahman P, Maksymowych WP, Xu H, Crusius JB, van der Horst-Bruinsma IE, Chou CT, Valle-Oñate R, Romero-Sánchez C, Hansen IM, Pimentel-Santos FM, Inman RD, Videm V, Martin J, Breban M, Reveille JD, Evans DM, Kim TH, Wordsworth BP, Brown MA. Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci. Nat Genet. 2013;45:730–8.PubMedPubMedCentralCrossRef Cortes A, Hadler J, Pointon JP, Robinson PC, Karaderi T, Leo P, Cremin K, Pryce K, Harris J, Lee S, Joo KB, Shim SC, Weisman M, Ward M, Zhou X, Garchon HJ, Chiocchia G, Nossent J, Lie BA, Førre Ø, Tuomilehto J, Laiho K, Jiang L, Liu Y, Wu X, Bradbury LA, Elewaut D, Burgos-Vargas R, Stebbings S, Appleton L, Farrah C, Lau J, Kenna TJ, Haroon N, Ferreira MA, Yang J, Mulero J, Fernandez-Sueiro JL, Gonzalez-Gay MA, Lopez-Larrea C, Deloukas P, Donnelly P, Australo-Anglo-American Spondyloarthritis Consortium (TASC); Groupe Française d'Etude Génétique des Spondylarthrites (GFEGS); Nord-Trøndelag Health Study (HUNT); Spondyloarthritis Research Consortium of Canada (SPARCC); Wellcome Trust Case Control Consortium 2 (WTCCC2), Bowness P, Gafney K, Gaston H, Gladman DD, Rahman P, Maksymowych WP, Xu H, Crusius JB, van der Horst-Bruinsma IE, Chou CT, Valle-Oñate R, Romero-Sánchez C, Hansen IM, Pimentel-Santos FM, Inman RD, Videm V, Martin J, Breban M, Reveille JD, Evans DM, Kim TH, Wordsworth BP, Brown MA. Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci. Nat Genet. 2013;45:730–8.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Al-Mossawi MH, Chen L, Fang H, Ridley A, de Wit J, Yager N, Hammitzsch A, Pulyakhina I, Fairfax BP, Simone D, Yi Y, Bandyopadhyay S, Doig K, Gundle R, Kendrick B, Powrie F, Knight JC, Bowness P. Unique transcriptome signatures and GM-CSF expression in lymphocytes from patients with spondyloarthritis. Nat Commun. 2017;8:1510.PubMedPubMedCentralCrossRef Al-Mossawi MH, Chen L, Fang H, Ridley A, de Wit J, Yager N, Hammitzsch A, Pulyakhina I, Fairfax BP, Simone D, Yi Y, Bandyopadhyay S, Doig K, Gundle R, Kendrick B, Powrie F, Knight JC, Bowness P. Unique transcriptome signatures and GM-CSF expression in lymphocytes from patients with spondyloarthritis. Nat Commun. 2017;8:1510.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Gaublomme JT, Yosef N, Lee Y, Gertner RS, Yang LV, Wu C, Pandolfi PP, Mak T, Satija R, Shalek AK, Kuchroo VK, Park H, Regev A. Single-Cell Genomics Unveils Critical Regulators of Th17 Cell Pathogenicity. Cell. 2015;163:1400–12.PubMedPubMedCentralCrossRef Gaublomme JT, Yosef N, Lee Y, Gertner RS, Yang LV, Wu C, Pandolfi PP, Mak T, Satija R, Shalek AK, Kuchroo VK, Park H, Regev A. Single-Cell Genomics Unveils Critical Regulators of Th17 Cell Pathogenicity. Cell. 2015;163:1400–12.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Hsieh WS, Kung CC, Huang SL, Lin SC, Sun WH. TDAG8, TRPV1, and ASIC3 involved in establishing hyperalgesic priming in experimental rheumatoid arthritis. Sci Rep. 2017;7:8870.PubMedPubMedCentralCrossRef Hsieh WS, Kung CC, Huang SL, Lin SC, Sun WH. TDAG8, TRPV1, and ASIC3 involved in establishing hyperalgesic priming in experimental rheumatoid arthritis. Sci Rep. 2017;7:8870.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Chen CL, Liu FL, Lee CC, Chen TC, Ahmed Ali AA, Sytwu HK, Chang DM, Huang HS. Modified salicylanilide and 3-phenyl-2H-benzo[e][1,3]oxazine-2,4(3H)-dione derivatives as novel inhibitors of osteoclast differentiation and bone resorption. J Med Chem. 2014;57:8072–85.PubMedCrossRef Chen CL, Liu FL, Lee CC, Chen TC, Ahmed Ali AA, Sytwu HK, Chang DM, Huang HS. Modified salicylanilide and 3-phenyl-2H-benzo[e][1,3]oxazine-2,4(3H)-dione derivatives as novel inhibitors of osteoclast differentiation and bone resorption. J Med Chem. 2014;57:8072–85.PubMedCrossRef
21.
Zurück zum Zitat Lee CC, Liu FL, Chen CL, Chen TC, Chang DM, Huang HS. Discovery of 5-(2',4'-difluorophenyl)-salicylanilides as new inhibitors of receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis. Eur J Med Chem. 2015;98:115–26.PubMedCrossRef Lee CC, Liu FL, Chen CL, Chen TC, Chang DM, Huang HS. Discovery of 5-(2',4'-difluorophenyl)-salicylanilides as new inhibitors of receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis. Eur J Med Chem. 2015;98:115–26.PubMedCrossRef
22.
Zurück zum Zitat Dowty ME, Jesson MI, Ghosh S, Lee J, Meyer DM, Krishnaswami S, Kishore N. Preclinical to clinical translation of tofacitinib, a Janus kinase inhibitor, in rheumatoid arthritis. J Pharmacol Exp Ther. 2014;348:165–73.PubMedCrossRef Dowty ME, Jesson MI, Ghosh S, Lee J, Meyer DM, Krishnaswami S, Kishore N. Preclinical to clinical translation of tofacitinib, a Janus kinase inhibitor, in rheumatoid arthritis. J Pharmacol Exp Ther. 2014;348:165–73.PubMedCrossRef
23.
Zurück zum Zitat Dai SP, Huang YH, Chang CJ, Huang YF, Hsieh WS, Tabata Y, Ishii S, Su WS. TDAG8 involved in initiating inflammatory hyperalgesia and establishing hyperalgesic priming in mice. Sci Rep. 2017;7:41415.PubMedPubMedCentralCrossRef Dai SP, Huang YH, Chang CJ, Huang YF, Hsieh WS, Tabata Y, Ishii S, Su WS. TDAG8 involved in initiating inflammatory hyperalgesia and establishing hyperalgesic priming in mice. Sci Rep. 2017;7:41415.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Su YS, Mei HR, Wang CH, Sun WH. Peripheral 5-HT3 mediates mirror-image pain by a cross-talk with acid-sensing ion channel 3. Neuropharmacology. 2018;130:92–104.PubMedCrossRef Su YS, Mei HR, Wang CH, Sun WH. Peripheral 5-HT3 mediates mirror-image pain by a cross-talk with acid-sensing ion channel 3. Neuropharmacology. 2018;130:92–104.PubMedCrossRef
25.
Zurück zum Zitat Chen YJ, Huang CW, Lin CS, Chang WH, Sun WH. Expression and function of proton-sensing G protein-coupled receptors in inflammatory pain. Mol Pain. 2009;5:39.PubMedPubMedCentralCrossRef Chen YJ, Huang CW, Lin CS, Chang WH, Sun WH. Expression and function of proton-sensing G protein-coupled receptors in inflammatory pain. Mol Pain. 2009;5:39.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Huang CW, Tzeng JN, Chen YJ, Tsai WF, Chen CC, Sun WH. Nociceptors of dorsal root ganglion express proton-sensing G-protein-coupled receptors. Mol Cell Neurosci. 2007;36:195–210.PubMedCrossRef Huang CW, Tzeng JN, Chen YJ, Tsai WF, Chen CC, Sun WH. Nociceptors of dorsal root ganglion express proton-sensing G-protein-coupled receptors. Mol Cell Neurosci. 2007;36:195–210.PubMedCrossRef
27.
Zurück zum Zitat Mogi C, Tobo M, Tomura H, Murata N, He XD, Sato K, Kimura T, Ishizuka T, Sasaki T, Sato T, Kihara Y, Ishii S, Harada A, Okajima F. Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J Immunol. 2009;182:3243–51.PubMedCrossRef Mogi C, Tobo M, Tomura H, Murata N, He XD, Sato K, Kimura T, Ishizuka T, Sasaki T, Sato T, Kihara Y, Ishii S, Harada A, Okajima F. Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J Immunol. 2009;182:3243–51.PubMedCrossRef
28.
Zurück zum Zitat Onozawa Y, Fujita Y, Kuwabara H, Nagasaki M, Komai T, Oda T. Activation of T cell death-associated gene 8 regulates the cytokine production of T cells and macrophages in vitro. Eur J Pharmacol. 2012;683:325–31.PubMedCrossRef Onozawa Y, Fujita Y, Kuwabara H, Nagasaki M, Komai T, Oda T. Activation of T cell death-associated gene 8 regulates the cytokine production of T cells and macrophages in vitro. Eur J Pharmacol. 2012;683:325–31.PubMedCrossRef
29.
Zurück zum Zitat Jin Y, Sato K, Tobo A, Mogi C, Tobo M, Murata N, Ishii S, Im DS, Okajima F. Inhibition of interleukin-1β production by extracellular acidification through the TDAG8/cAMP pathway in mouse microglia. J Neurochem. 2014;129:683–95.PubMedCrossRef Jin Y, Sato K, Tobo A, Mogi C, Tobo M, Murata N, Ishii S, Im DS, Okajima F. Inhibition of interleukin-1β production by extracellular acidification through the TDAG8/cAMP pathway in mouse microglia. J Neurochem. 2014;129:683–95.PubMedCrossRef
30.
Zurück zum Zitat Wang JQ, Kon J, Mogi C, Tobo M, Damirin A, Sato K, Komachi M, Malchinkhuu E, Murata N, Kimura T, Kuwabara A, Wakamatsu K, Koizumi H, Uede T, Tsujimoto G, Kurose H, Sato T, Harada A, Misawa N, Tomura H, Okajima F. TDAG8 is a proton-sensing and psychosine-sensitive G-protein-coupled receptor. J Biol Chem. 2004;279:45626–33.PubMedCrossRef Wang JQ, Kon J, Mogi C, Tobo M, Damirin A, Sato K, Komachi M, Malchinkhuu E, Murata N, Kimura T, Kuwabara A, Wakamatsu K, Koizumi H, Uede T, Tsujimoto G, Kurose H, Sato T, Harada A, Misawa N, Tomura H, Okajima F. TDAG8 is a proton-sensing and psychosine-sensitive G-protein-coupled receptor. J Biol Chem. 2004;279:45626–33.PubMedCrossRef
31.
Zurück zum Zitat Ishii S, Kihara Y, Shimizu T. Identification of T cell death-associated gene 8 (TDAG8) as a novel acid sensing G-protein coupled receptor. J Biol Chem. 2005;280:9083–7.PubMedCrossRef Ishii S, Kihara Y, Shimizu T. Identification of T cell death-associated gene 8 (TDAG8) as a novel acid sensing G-protein coupled receptor. J Biol Chem. 2005;280:9083–7.PubMedCrossRef
32.
Zurück zum Zitat Richter F, Natura G, Ebbinghaus M, von Banchet GS, Hensellek S, König C, Bräuer R, Schaible HG. Interleukin-17 sensitizes joint nociceptors to mechanical stimuli and contributes to arthritic pain through neuronal interleukin-17 receptors in rodents. Arthritis Rheum. 2012;64:4125–34.PubMedCrossRef Richter F, Natura G, Ebbinghaus M, von Banchet GS, Hensellek S, König C, Bräuer R, Schaible HG. Interleukin-17 sensitizes joint nociceptors to mechanical stimuli and contributes to arthritic pain through neuronal interleukin-17 receptors in rodents. Arthritis Rheum. 2012;64:4125–34.PubMedCrossRef
33.
Zurück zum Zitat Ebbinghaus M, Segond von Banchet G, Massier J, Gajda M, Bräuer R, Kress M, Schaible HG. Interleukin-6-dependent influence of nociceptive sensory neurons on antigen-induced arthritis. Arthritis Res Ther. 2015;17:334.PubMedPubMedCentralCrossRef Ebbinghaus M, Segond von Banchet G, Massier J, Gajda M, Bräuer R, Kress M, Schaible HG. Interleukin-6-dependent influence of nociceptive sensory neurons on antigen-induced arthritis. Arthritis Res Ther. 2015;17:334.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Ebbinghaus M, Natura G, Segond von Banchet G, Hensellek S, Böttcher M, Hoffmann B, Salah FS, Gajda M, Kamradt T, Schaible HG. Interleukin-17A is involved in mechanical hyperalgesia but not in the severity of murine antigen-induced arthritis. Sci Rep. 2017;7:10334.PubMedPubMedCentralCrossRef Ebbinghaus M, Natura G, Segond von Banchet G, Hensellek S, Böttcher M, Hoffmann B, Salah FS, Gajda M, Kamradt T, Schaible HG. Interleukin-17A is involved in mechanical hyperalgesia but not in the severity of murine antigen-induced arthritis. Sci Rep. 2017;7:10334.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Peng J, Gu N, Zhou L, Eyo UB, Murugan M, Gan WB, Wu LL. Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury. Nat Commun. 2016;7:12029.PubMedPubMedCentralCrossRef Peng J, Gu N, Zhou L, Eyo UB, Murugan M, Gan WB, Wu LL. Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury. Nat Commun. 2016;7:12029.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Costa FA, Moreira Neto FL. Satellite glial cells in sensory ganglia: its role in pain. Rev Bras Anestesiol. 2015;65:73–81.PubMedCrossRef Costa FA, Moreira Neto FL. Satellite glial cells in sensory ganglia: its role in pain. Rev Bras Anestesiol. 2015;65:73–81.PubMedCrossRef
37.
Zurück zum Zitat Onozawa Y, Komai T, Oda T. Activation of T cell death-associated gene 8 attenuates inflammation by negatively regulating the function of inflammatory cells. Eur J Pharmacol. 2011;654:315–9.PubMedCrossRef Onozawa Y, Komai T, Oda T. Activation of T cell death-associated gene 8 attenuates inflammation by negatively regulating the function of inflammatory cells. Eur J Pharmacol. 2011;654:315–9.PubMedCrossRef
38.
Zurück zum Zitat Davignon JL, Hayder M, Baron M, Boyer JF, Constantin A, Apparailly F, Poupot R, Cantagrel A. Targeting monocytes/macrophages in the treatment of rheumatoid arthritis. Rheumatology. 2013;52:590–8.PubMedCrossRef Davignon JL, Hayder M, Baron M, Boyer JF, Constantin A, Apparailly F, Poupot R, Cantagrel A. Targeting monocytes/macrophages in the treatment of rheumatoid arthritis. Rheumatology. 2013;52:590–8.PubMedCrossRef
39.
Zurück zum Zitat Donlin LT, Jayatilleke A, Giannopoulou EG, Kalliolias GD, Ivashkiv LB. Modulation of TNF-induced macrophage polarization by synovial fibroblast. J Immunol. 2014;193:2373–83.PubMedCrossRef Donlin LT, Jayatilleke A, Giannopoulou EG, Kalliolias GD, Ivashkiv LB. Modulation of TNF-induced macrophage polarization by synovial fibroblast. J Immunol. 2014;193:2373–83.PubMedCrossRef
40.
Zurück zum Zitat Redlich K, Hayer S, Ricci R, David JP, Tohidast-Akrad M, Kollias G, Steiner G, Smolen JS, Wagner EF, Schett G. Osteoclasts are essential for TNF-α-mediated joint destruction. J Clin Invest. 2002;110:1419–27.PubMedPubMedCentralCrossRef Redlich K, Hayer S, Ricci R, David JP, Tohidast-Akrad M, Kollias G, Steiner G, Smolen JS, Wagner EF, Schett G. Osteoclasts are essential for TNF-α-mediated joint destruction. J Clin Invest. 2002;110:1419–27.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Williams EK, Chang RB, Strochlic DE, Umans BD, Lowell BB, Liberles SD. Sensory Neurons that Detect Stretch and Nutrients in the Digestive System. Cell. 2016;166:209–21.PubMedPubMedCentralCrossRef Williams EK, Chang RB, Strochlic DE, Umans BD, Lowell BB, Liberles SD. Sensory Neurons that Detect Stretch and Nutrients in the Digestive System. Cell. 2016;166:209–21.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Wu W, Liu HP, Chen F, Liu H, Cao AT, Yao S, Sun M, Evans-Marin HL, Zhao Y, Zhao Q, Duck LW, Elson CO, Liu Z, Cong Y. Commensal A4 bacteria inhibit intestinal Th2-cell responses through induction of dendritic cell TGF-β production. Eur J Immunol. 2016;46:1162–7.PubMedPubMedCentralCrossRef Wu W, Liu HP, Chen F, Liu H, Cao AT, Yao S, Sun M, Evans-Marin HL, Zhao Y, Zhao Q, Duck LW, Elson CO, Liu Z, Cong Y. Commensal A4 bacteria inhibit intestinal Th2-cell responses through induction of dendritic cell TGF-β production. Eur J Immunol. 2016;46:1162–7.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Hu W, Chen FH, Yuan FL, Zhang TY, Wu FR, Rong C, Jiang S, Tang J, Zhang CC, Lin MY. Blockade of acid-sensing ion channels protects articular chondrocytes from acid-induced apoptotic injury. Inflamm Res. 2012;61:327–35.PubMedCrossRef Hu W, Chen FH, Yuan FL, Zhang TY, Wu FR, Rong C, Jiang S, Tang J, Zhang CC, Lin MY. Blockade of acid-sensing ion channels protects articular chondrocytes from acid-induced apoptotic injury. Inflamm Res. 2012;61:327–35.PubMedCrossRef
Metadaten
Titel
TDAG8 deficiency reduces satellite glial number and pro-inflammatory macrophage number to relieve rheumatoid arthritis disease severity and chronic pain
verfasst von
Shih-Ping Dai
Wei-Shan Hsieh
Chien-Hua Chen
Yueh-Hao Lu
Hsu-Shan Huang
Der-Ming Chang
Shir-Ly Huang
Wei-Hsin Sun
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2020
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-020-01851-z

Weitere Artikel der Ausgabe 1/2020

Journal of Neuroinflammation 1/2020 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.