Open Access

Activation of TGR5 alleviates inflammation in rheumatoid arthritis peripheral blood mononuclear cells and in mice with collagen II‑induced arthritis

  • Authors:
    • Zhe‑Yong Li
    • Jing‑Jing Zhou
    • Chun‑Lei Luo
    • Le‑Meng Zhang
  • View Affiliations

  • Published online on: September 26, 2019     https://doi.org/10.3892/mmr.2019.10711
  • Pages: 4540-4550
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rheumatoid arthritis (RA) is characterized by chronic inflammatory synovitis resulting in progressive joint destruction. Persistent synovial inflammation is induced by activation of various inflammatory cells. G‑protein‑coupled bile acid receptor 1 (TGR5) is a G‑protein‑coupled receptor activated by various bile acids, which has been reported to act as a key adaptor in regulating various signaling pathways involved in inflammatory responses and a diverse array of physiological processes, including bile acid synthesis, lipid and carbohydrate metabolism, carcinogenesis, immunity and inflammation. In the present study, TGR5 expression was detected in RA peripheral blood mononuclear cells (PBMCs), and its association with clinical disease activity, histological synovitis severity and radiological joint destruction was analyzed. Subsequently, the role and potential underlying mechanisms of TGR5 in the PBMCs of patients with RA and mice with collagen II‑induced arthritis (CIA) were investigated. PBMCs were obtained from 50 patients with RA and 40 healthy controls (HCs). The mRNA and protein expression levels of TGR5 were detected in PBMCs via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and immunofluorescence staining, respectively. Additionally, the levels of proinflammatory cytokines were analyzed by RT‑qPCR and enzyme‑linked immunosorbent assay (ELISA). The activation of nuclear factor‑κB (NF‑κB) and IκB kinase a was determined via western blot analysis. The anti‑arthritic and anti‑inflammatory effects of LCA on mice with CIA were then investigated. The arthritis score was assessed, and the protein levels of proinflammatory cytokines in the plasma of mice were detected via ELISA. TGR5 mRNA expression was significantly downregulated in the PBMCs of patients with RA compared with in those of the HCs (0.53±0.58 for patients vs. 1.49±0.83 for HCs; P<0.001); similar findings were observed at the protein level. The mRNA expression levels of TGR5 in the PBMCs of patients with RA with a high 28‑Joint Disease Activity Score (DAS28) were significantly decreased compared with in patients with a low DAS28 (0.81±0.65 for low score vs. 0.35±0.46 for high score; P=0.002). Furthermore, TGR5 expression was significantly correlated with the levels of C‑reactive protein (r=‑0.429; P=0.002) and the DAS28 (r=‑0.383; P=0.006). RT‑qPCR and ELISA analyses indicated that lithocholic acid (LCA, 10 mg/kg/day) attenuated lipopolysaccharide‑induced proinflammatory cytokine production via inhibition of NF‑κB activity in the PBMCs of patients with RA. In addition, the arthritis score was significantly decreased in LCA‑treated CIA mice compared with in non‑treated CIA mice. The increased production of tumor necrosis factor‑α, interleukin (IL)‑1β, IL‑6 and IL‑8 was significantly reduced in the plasma of LCA‑treated CIA mice compared with the control. In conclusion, TGR5 may contribute to the inflammation of PBMCs in patients with RA and mice with CIA.

Introduction

Rheumatoid arthritis (RA) is characterized by chronic inflammatory synovitis resulting in progressive joint destruction (1). Persistent synovial inflammation is considered to be a characteristic feature of RA, leading and contributing to cartilage and bone destruction, and subsequent disability in RA (2); however, there is no effective treatment for RA, and the precise etiology and underlying mechanisms remain unclear. At present, various cell types have been reported to serve distinct, complex and interconnected roles in the synovial inflammation of RA, including monocytes/macrophages, T cells, B cells, fibroblast-like synoviocytes (FLSs) and osteoclasts (3,4). For example, FLSs in the intimal lining of the synovium serve an important role in RA via the hyperproduction of proinflammatory cytokines and matrix metalloproteinases (MMPs), potentially resulting in cartilage destruction (5).

Monocytes/macrophages are recognized as a prominent joint-specific determinant; their activation may lead to inflammation, acute phase reaction and joint destruction in acute and chronic RA by promoting the production of key proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and MMPs (68). In addition, these cells are a potent source of chemokines, which recruit additional leukocytes to the inflamed joint to exacerbate the disease process. Furthermore, monocytes can differentiate into osteoclasts, and have also been reported to enhance the production of the osteoclastogenic cytokine IL-17 by CD4+ T cells, in turn further contributing to bone erosion, and focal and systemic osteoporosis (6,9,10). The importance of the role of macrophages in RA is supported by findings reporting these cells to be a reliable biomarker for treatment response and associated with disease activity markers (1113). Nuclear factor-κB (NF-κB) has received considerable attention as a key regulator of immunology, inflammation and cancer development, and has been reported to influence the pathogenesis and progression of RA (14,15). Activation of NF-κB promotes the secretion of cytokines such as TNF-α and IL-1β by the monocytes/macrophages of patients with RA (16). Therefore, increasing attention in the field of RA research has been placed on monocytes/macrophages as potential therapeutic targets (7).

G-protein-coupled bile acid receptor 1 (TGR5) is a G-protein-coupled receptor, which responds to various bile acids by activating transcriptional networks and/or signaling cascades, which regulate a diverse array of physiological processes, including bile acid synthesis, lipid and carbohydrate metabolism, carcinogenesis and inflammation (17,18). TGR5 gene expression is diffusely distributed in the endocrine glands, muscles, immune organs, adipocytes, spinal cord and enteric nervous system (19). Human and mouse TGR5 exhibit similar affinity for the different bile acid species; the highest affinity is for lithocholic acid (LCA), followed by deoxycholic acid, chenodeoxycholic acid and cholic acid (20,21). Since the identification of dedicated bile acid receptors, there has been an increased interest in modulating these targets pharmacologically. For example, the observation that TGR5 induces the secretion of clinically relevant glucagon-like peptide 1 has promoted the development of a novel class of drugs for the treatment of type 2 diabetes mellitus (22). Regarding the potential for manipulating TGR5 within the context of immunology and inflammation, studies on rabbit alveolar macrophages and human hepatic Kupffer cells have suggested that TGR5 activation suppresses lipopolysaccharide (LPS)-induced production of the proinflammatory cytokines TNF-α, IL-1, IL-6 and IL-8 (23,24). The potential for targeting TGR5 in immunology has been further validated by the finding that TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading (25). Furthermore, previous studies on mouse bone marrow-derived macrophages and human macrophage subsets revealed that TGR5 activation negatively regulates hepatic inflammatory responses via impaired NF-κB signaling pathways (26,27).

RA is one of the most severe chronic diseases, which is caused by persistent synovial inflammation and bone destruction. TGR5 is important for inflammatory signaling pathways; however, at present, the expression and pathophysiological role of TGR5 in RA have not been elaborated. Therefore, in the present study, TGR5 expression in RA peripheral blood mononuclear cells (PBMCs) was determined, and its association with clinical disease activity, histological synovitis severity and radiological joint destruction was investigated. Subsequently, the role of TGR5 in PBMC inflammation and the potential mechanisms underlying its effects were determined. Finally, the anti-arthritic and anti-inflammatory effects of LCA on mice with collagen type II (CII)-induced arthritis (CIA) were investigated.

Materials and methods

Patients

A total of 50 Chinese patients with RA who fulfilled the 1987 revised criteria of the American College of Rheumatology (ACR) (28) or the 2010 ACR/European League against Rheumatism classification criteria for RA (29) were recruited from the Department of Rheumatology and Orthopedics of the Hunan Cancer Hospital (Changsha, China). All patients presented with active disease, described as the presence of a 28-Joint Disease Activity Score (DAS28) of ≥3.2. Age, gender or disease duration did not differ among the patients with RA and healthy controls. In total, 11 RA patients were male and 39 were female, with a mean age of 59 (49–64) years. In the healthy control group, 14 patients were male and 26 were female, with a mean age of 36 (2948) years. The date range of assessment/sample collection for both groups was from June 2016 to October 2018. The present study was conducted in compliance with the Helsinki Declaration principles. All patients provided written informed consent

Clinical assessments

The clinical data of all patients with RA were collected at baseline, including the 28-joint tender and swollen joint counts (28TJC and 28SJC, respectively), patient and provider global assessment of disease activity (PtGA and PrGA, respectively) scores, visual analog scale score for pain, Chinese language version of the Stanford Health Assessment Questionnaire (HAQ) score (30), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP) level, rheumatoid factor (RF) level and anti-cyclic citrullinated peptide (anti-CCP) antibody level. Disease activity was assessed using the DAS28 with four variables, including CRP level [DAS28 (4)-CRP] (31).

Isolation and culture of human PBMCs

Heparinized whole blood was collected from 50 patients with RA and 40 healthy controls (HCs). PBMCs were separated via Ficoll/Paque density gradient centrifugation at 400 × g for 30 min at 18–20°C. Cells were then re-suspended in α-Minimal Essential Medium with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and plated in a 37°C humidified incubator with 5% CO2. On the following day, the suspended cells were removed and washed thoroughly, and the adherent cells were collected and plated. PBMCs were first treated with various concentrations (0, 50 and 100 µM) of LCA for 60 min, and then treated with LPS (100 ng/ml) for 12 h, mRNA was extracted, and the culture medium was collected for ELISA analysis.

Immunofluorescence staining

The PBMCs (8×105 cells/well) were plated on 24-well culture plates with coverslips, fixed in 4% paraformaldehyde for 20 min at room temperature, permeabilized with 0.2% Triton X-100 for 10 min at room temperature, and blocked in PBS containing 3% bovine serum albumin (Abcam) for 30 min at 37°C. The cells were then incubated in PBS containing rabbit anti-human polyclonal antibody against TGR5 (10 µg/ml; ab72608; Abcam) overnight at 4°C, and subsequently incubated with Alexa Fluor® 633-conjugated goat anti-rabbit immunoglobulin G secondary antibodies (1:1,000; cat. no. A-21071; Invitrogen; Thermo Fisher Scientific, Inc.) for 1 h at 37°C. Subsequently, DAPI (Sigma-Aldrich; Merck KGaA) was used to stain the cell nuclei at a concentration of 1.43 µM for 3 min at room temperature, and cells were mounted using ProLong® Gold Antifade Reagent (cat. no. P36934; Invitrogen; Thermo Fisher Scientific, Inc.). The images were acquired using a 160 Zeiss LSM 510 Confocal Imaging system (Zeiss GmbH).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from PBMCs using the RNAiso Plus reagent (Takara Bio, Inc.) according to the manufacturer's protocols. First-strand complementary DNA (cDNA) was synthesized from each total RNA sample using a PrimeScript 1st Strand cDNA Synthesis kit (Takara Bio, Inc.) at 37°C for 15 min, 85°C for 5 sec. qPCR was performed using the QuantiTect™ SYBR Green PCR kit (Takara Bio, Inc.) on the Roche LightCycler480 sequence detector system (Roche Diagnostics) according to the manufacturer's protocols. The qPCR conditions were as follows: Denaturation at 95°C for 30 sec, then 95°C for 5 sec and 60°C for 30 sec for 40–50 cycles. The sequences of the primers used in the study are presented in Table I, and these were synthesized by Invitrogen (Thermo Fisher Scientific, Inc.). PCR amplification of the housekeeping gene β-actin was performed for each sample as a control for sample loading. Normalization and quantification of the PCR signals was performed by comparing the cycle threshold value of the gene of interest, in triplicate, with β-actin. The fold change in gene expression relative to the control was calculated using the 2−ΔΔCq method (32). Data are presented as the mean ± standard deviation of three independent experiments and analyzed with the LightCycler480 software release 1.5.0 (Roche Diagnostics).

Table I.

Primers for reverse transcription-quantitative polymerase chain reaction.

Table I.

Primers for reverse transcription-quantitative polymerase chain reaction.

GeneSequence
β-actinSense: 5′-GGACTTCGAGCAAGAGATGG-3′
Antisense: 5′-TGTGTTGGCGTACAGGTCTTTG-3′
IL-6Sense: 5′-CTGCGCAGCTTTAAGGAGTTC-3′
Antisense: 5′-CAATCTGAGGTGCCCATGCTA-3′
IL-8Sense: 5′-GTGCAGAGGGTTGTGGAGAAGTTT-3′
Antisense: 5′-TCACTGGCATCTTCACTGATTCTTG-3′
IL-1βSense: 5′-CCAGCTACGAATCTCCGACC-3′
Antisense: 5′-CATGGCCACAACAACTGACG-3′
TNF-aSense: 5′-GCTAAGAGGGAGAGAAGCAACTACA-3′
Antisense: 5′-GAAGAGGCTGAGGAACAAGCA-3′
TGR5Sense: 5′-CCCAGGCTATCTTCCCAGC-3′
Antisense: 5′-GCCAGGACTGAGAGGAGCA-3′

[i] IL, interleukin; TGR5, G-protein-coupled bile acid receptor 1; TNF-α, tumor necrosis factor-α.

Western blot analysis

PBMCs were collected and lysed to obtain proteins using a RIPA buffer (Cell Signaling Technologies, Inc.), whereas nuclear protein was separated using a NE-PER™ Nuclear and Cytoplasmic Extraction kit (Pierce; Thermo Fisher Scientific, Inc.). Protein concentration was measured using bicinchoninic acid Protein Assay kit from Bio-Rad Laboratories, Inc. Proteins (50 µg/lane) were separated by 12% SDS-PAGE under denaturing conditions, and were electrotransferred to nitrocellulose membranes. The membranes were then blocked with 5% non-fat milk in 0.1% TBS-Tween 20 for 1 h at room temperature, and were probed with antibodies against TGR5 (cat. no. ab72608; Abcam), phosphorylated (p)-IκB kinase α (IκBα; cat. no. 2859; Cell Signaling Technologies, Inc.), total (T)-IκBα (cat. no. 9249; Cell Signaling Technologies, Inc.) and p-NF-κB (cat. no. 3033; Cell Signaling Technologies, Inc.; all 1:1,000); GAPDH (cat. no. 5174; Cell Signaling Technologies, Inc.; 1:1,000) and histone H3 (cat. no. 9275; Cell Signaling Technologies, Inc.; 1:2,000) overnight at 4°C. For the detection of proteins, membranes were then incubated for 1 h at room temperature with horseradish peroxidase-conjugated secondary antibody (cat. no. E030120-01; EarthOx Life Sciences; 1:5,000). Immunoreactive bands were visualized using an enhanced chemiluminescence reagent (EMD Millipore). Protein bands were semi-quantified via densitometric analyses using a G:BOX Gel & Blot Imaging Series (Syngene Europe) and ImageQuant LAS500 (GE Life Sciences). Phosphorylated protein expression was normalized to total protein expression, and expressed as a fold change relative to the untreated control. Data are presented as the means ± standard deviation of three independent experiments.

Enzyme-linked immunosorbent assay (ELISA)

The quantities of TNF-α (cat. no. DTA00D, R&D Systems, Inc.), IL-1β (cat. no. DLB50, R&D Systems, Inc.), IL-6 (cat. no. D6050, R&D Systems, Inc.) and IL-8 (cat. no. D8000C, R&D Systems, Inc.) in culture supernatants were measured via ELISA according to the manufacturer's protocols. The quantities of TNF-α (cat. no. MTA00B, R&D Systems, Inc.), IL-1β (cat. no. MLB00C, R&D Systems, Inc.), IL-6 (cat. no. M6000B, R&D Systems, Inc.) and IL-8 (cat. no. LS-F55769, LifeSpan BioSciences, Inc.) in mouse blood samples were measured via ELISA according to the manufacturer's protocol. Briefly, PBMCs were subjected to different treatments. Then, mouse blood samples were centrifuged at 1,006.2 × g for 15 min to separate the blood plasma at room temperature. The culture supernatant or the blood plasma were mixed with the assay buffer and added to anti-TNF-α, IL-1β, IL-6 and IL-8 antibody-coated wells. Horseradish peroxidase-conjugated anti-human TNF-α, IL-1β, IL-6, and IL-8 monoclonal antibodies were added and incubated at 37°C for 2 h, followed by incubation with colorimetric (tetramethylbenzidine) solution for a further 10 min. The relative absorbance was measured at 450 nm, and three independent experiments were performed for each condition.

Animal study

This study was conducted on male DAB/1J mice (age, 6–8 weeks; 18–22 g) obtained from Beijing HFK Bioscience Co., Ltd. A total of 30 mice were divided into three equal groups: Negative controls (NCs), experimental arthritis controls not treated with LCA and experimental arthritis group treated with LCA (10 mg/kg/day; Sigma-Aldrich; Merck KGaA). The NCs did not have CIA. The mice were housed in a room on a 12-h light/dark cycle under specific pathogen-free conditions, at a temperature of 22–26°C and relative humidity of 55–65%. All experiments were conducted in compliance with the National Institutes of Health guidelines for the care and use of laboratory animals (33). The study was approved by the Institutional Animal and Clinical Committees of Hunan Cancer Hospital.

Induction of CIA and LCA treatment

CIA was induced according to the previously described method by Brand et al (34) in the experimental arthritis controls not treated with LCA and experimental arthritis group treated with LCA mouse groups. Briefly, native chick CII (Chondrex, Inc.) was dissolved in 10 mM acetic acid to a 1 mg/ml concentration, and was emulsified in an equal volume of Freund's complete adjuvant (Sigma-Aldrich; Merck KGaA). Each mouse received two intradermal injections of emulsion (0.1 ml) containing CII (1 mg/ml) and Freund's complete adjuvant at two separated sites tail on days 1 and 21.

For LCA treatment, LCA was dissolved and freshly diluted in PBS. PBS and LCA (10 mg/kg/day) were orally administered via a gastric tube once a day from day 21 to day 42 following the first immunization. Both NCs and experimental arthritis controls not treated with LCA were treated with PBS.

Arthritis score

The arthritis score was assessed once every 3 days following the initiation of LCA treatment. The severity of arthritis was measured in a double manner using the semi-quantitative clinical scoring system described by Yoo et al (35) (0, no arthritis; 1, definite erythema and swelling of the ankle or one digit; 2, two or more joints involved or mild erythema and swelling of the entire paw; 3, erythema and swelling extending from the ankle to the metatarsal joints of the entire paw and all digits; and 4, ankylosing deformity with severe joint erythema and swelling). The arthritis score for each mouse was the sum of all paw scores (yielding a score between 0 and 16). On day 42, the mice were sacrificed and blood was collected for serum separation.

Statistical analysis

Statistical analyses were performed using SPSS 13.0 software (SPSS, Inc.). For categorical variables, data are presented as frequencies and percentages. For continuous variables, data are presented as the means ± standard deviation, or the median and interquartile range. Wilcoxon rank-sum test was used to compare the expression of TGR5 in the PBMCs of patients with RA and HCs, and the arthritis score in the experimental arthritis controls and experimental arthritis controls treated with LCA. One-way or Welch's ANOVA followed by least significant difference post-hoc tests were performed to compare the expression of TNF-α, IL-1β, IL-6 and IL-8 in the culture supernatant or the blood plasma, and the expression of p-NF-κB and p-IκBα in the PBMCs treated with various LCA or not treated with LCA. Spearman's rank order correlation test was used to assess the correlation between TGR5 expression in RA PBMCs and clinical parameters. P<0.05 was considered to indicate a statistically significant difference.

Results

Characteristics of the study patients

Table II presents the baseline demographic and clinical features of individuals in the study. The gender ratio did not differ between the patients in the RA and HC groups. Among the patients with RA, 48% (24/50) had not previously taken corticosteroids or disease-modifying anti-rheumatic drugs (DMARDs). The majority of patients had taken only Chinese herbal medicine and/or painkillers to relieve arthralgia. At recruitment, 18% (9/50) had taken corticosteroids alone. The remaining 34% (17/50) received treatment with one or more DMARD, including methotrexate, leflunomide, sulfasalazine, hydroxychloroquine and etanercept.

Table II.

Baseline demographic and clinical features of patients with RA and HCs.

Table II.

Baseline demographic and clinical features of patients with RA and HCs.

CharacteristicPatients with RA (n=50)HC (n=40)
Demographic
  Age [years; median (IQR)]59 (49–64)36 (29–48)
  Female [n (%)]39 (78)26 (65)
Disease status
  Disease duration [months; median (IQR)]34 (12–106)NA
  ESR [mm/h; median (IQR)]75 (54–105)11 (3–21)
  CRP [mg/dl; median (IQR)]4.02 (1.19–5.75)0.11 (0.02–0.23)
  Rheumatoid factor-positive [n (%)]47 (94)NA
  Anti-CCP-positive [n (%)]43 (86)NA
  DAS28 [median (IQR)]5.5 (4.6~6.3)NA
Previous medications [n (%)]
  Corticosteroids22 (50)NA
  Methotrexate20 (40)NA
  Leflunomide5 (10)NA
  Sulfasalazine4 (8)NA
  Hydroxychloroquine6 (12)NA
  Etanercept2 (4)NA

[i] Anti-CCP, anti-cyclic citrullinated peptide antibodies; CRP, C-reactive protein; DAS28, Disease Activity Score 28-joint assessment; ESR, erythrocyte sedimentation rate; HC, healthy control; IQR, interquartile range; NA, not applicable; RA, rheumatoid arthritis; RF, rheumatoid factor.

Expression of TGR5 is downregulated in the PBMCs of patients with RA

The mRNA expression levels of TGR5 in PBMCs were evaluated in 50 patients with RA and 40 HCs. The expression of TGR5 mRNA in the PBMCs of patients with RA was significantly decreased compared with in the HCs (0.53±0.58 vs. 1.49±0.83; P<0.001; Fig. 1A). Additionally, the expression of TGR5 mRNA in the PBMCs of patients with RA with a high DAS28 was significantly decreased compared with in patients with a low DAS28 (0.35±0.46 vs. 0.81±0.65, respectively; P=0.002; Fig. 1B). The expression of TGR5 mRNA in the PBMCs of the patients with RA with either a high or low DAS28 was also significantly lower than that of the HCs (0.81±0.65 vs. 1.49±0.83, P<0.001; 0.35±0.46 vs. 1.49±0.83, P=0.002; Fig. 1B). The number of patients with RA with a DAS28 score of ≥5.1 was 30. Of these patients, 50% (15/30) had never taken corticosteroids or DMARDs. At recruitment, 20% (6/30) had taken corticosteroids alone. The remaining 30% (9/30) received treatment with one or more DMARDs, including methotrexate, leflunomide, sulfasalazine, hydroxychloroquine or etanercept. Patients did not exhibit any obvious side effects after taking DMARDs. Immunofluorescence staining revealed markedly reduced expression of TGR5 in the PBMCs of patients with RA compared with in the PBMCs of HCs (Fig. 1C).

Expression of TGR5 mRNA in the PBMCs of patients with RA demonstrates a negative correlation with clinical parameters

Spearman's rank order correlation test revealed significant correlations between the mRNA expression levels of TGR5 in PBMCs and the CRP level (r=−0.429, P=0.002), and the DAS28 (r=−0.383, P=0.006) of patients (Fig. 2). There was no significant correlation between TGR5 mRNA expression and 28TJC, 28SJC, PtGA score, PrGA score, HAQ score, RF level, anti-CCP level, ESR, the simplified disease activity index and the clinical disease activity index (respectively: r=0.043, P=0.763; r=−0.072 P=0.611; r=−0.011, P=0.939; r=0.185, P=0.188; r=0.172, P=0.223; r=0.049, P=0.732; r=0.188, P=0182; r=0.067, P=0.642; r=−0.099, P=0.544; and r=−0.106, P=0.455).

LCA attenuates LPS-induced proinflammatory cytokine production in the PBMCs of patients with RA

LPS is a potent endotoxin involved in the progression of RA diseases (36). LPS treatment (100 ng/ml) for 12 h markedly increased the mRNA and protein expression levels of TNF-α, IL-1β, IL-6 and IL-8 in the PBMCs of patients with RA by between 3- and 10-fold (Fig. 3). Pretreatment of PBMCs with LCA (50 and 100 µM) for 60 min significantly inhibited LPS-induced TNF-α, IL-1β, IL-6 and IL-8 mRNA expression and protein release into the supernatant in a concentration-dependent manner (Fig. 3).

LCA inhibits LPS-induced NF-κB and IκBα activation in the PBMCs of patients with RA

To further investigate the mechanism underlying the effects of LCA on the inhibition of LPS-induced proinflammatory cytokine production, NF-κB and IκBα signaling was measured via western blot analysis. LCA treatment significantly decreased the phosphorylation of NF-κB and IκBα, but not the total protein levels of IκBα (Fig. 4).

Effects of LCA on DAB/1J mice with CIA

Mice with CIA have been frequently utilized to investigate arthritic diseases, as they possess various pathological features similar to those of human RA (37). There was no macroscopic evidence of paw erythema or edema in the normal blank control mice. As presented in Fig. 5A, arthritis symptoms in the mice with CIA progressed and maintained a high intensity from day 21 to day 42 following immunization; this was notably relieved following treatment with LCA (10 mg/kg/day). From day 30, the arthritis score was significantly decreased in the LCA treatment group compared with in the CIA model group (P<0.05 or 0.01).

To demonstrate its anti-inflammatory effects in vivo, LCA was evaluated for its inhibition of the production of proinflammatory cytokines in mice with CIA. Cytokines, such as TNF-α, IL-1β, IL-6 and IL-8, are associated with the progression and severity of arthritis (16). On day 42, the mice were sacrificed, and blood was collected for serum separation and subsequent ELISA detection. As presented in Fig. 5B, the serum levels of the proinflammatory cytokines TNF-α, IL-1β, IL-6 and IL-8 were significantly increased in the non-treated CIA mice compared with in the non-arthritic group. The increased TNF-α, IL-1β, IL-6, and IL-8 production was significantly suppressed by LCA treatment compared with in the non-treated mice with CIA.

Discussion

RA is an inflammatory disease characterized by chronic inflammatory synovitis resulting in progressive joint destruction. The pathogenesis of RA involves a diverse range of cells, including monocytes/macrophages, T cells, B cells and FLSs, which produce a large number of proinflammatory cytokines, MMPs and other proteolytic enzymes. In addition, previous studies have indicated that the presence of inflammatory cells is positively correlated with synovitis score, further demonstrating the involvement of inflammatory cells in the pathophysiology of RA (38,39).

Monocytic/macrophagic inflammation is central to almost all aspects that contribute to the development of RA, as it is regarded as the primary source of proinflammatory cytokines, including TNF-α and IL-1β, which serve important roles in the communication between the various inflammatory cells involved in RA (40,41). TGR5 has been identified as an important component of the bile acid signaling network, and its activation has been reported to control numerous physiological pathways associated with metabolic homeostasis, bile acid synthesis and inflammation. Furthermore, emerging evidence has demonstrated that TGR5 is expressed in various immune cells, including monocytes, alveolar macrophages and Kupffer cells, and it has been reported that its activation modulates inflammatory responses in these cells (23,24,26); however, whether TGR5 serves a role in the regulation of PBMC inflammation in RA has yet to be fully investigated. In the present study, it was revealed that TGR5 was expressed in primary PBMCs, and that TGR5 expression was significantly downregulated in the PBMCs of patients with RA compared with in PBMCs from HCs. Additionally, TGR5 expression in PBMCs exhibited significantly negative correlations with CRP level and DAS28. Therefore, it was hypothesized that decreased TGR5 expression may potentially be involved in the pathogenesis of RA.

To investigate the effects of TGR5 on PBMC-mediated inflammation, LPS was used to induce inflammation in PBMCs, and LCA was administered to activate TGR5. It was observed that pretreatment of PBMCs with LCA potently downregulated LPS-induced TNF-α, IL-1β, IL-6 and IL-8 expression, and inhibited NF-κB and IκBα phosphorylation in a concentration-dependent manner; this finding is consistent with previous observations demonstrating that TGR5 activation attenuates proinflammatory cytokine production in monocytes/macrophages via suppression of the nuclear translocation of NF-κB (23,27,42). Högenauer et al (42) indicated that TGR5 activation inhibits LPS-induced TNF-α and IL-12 release from PBMCs and in mice. Additionally, it has been reported that TGR5 activation does not affect production of the anti-inflammatory cytokine IL-10, it only suppresses the secretion of proinflammatory cytokines TNF-α, IL-6 and IL-12p40 by primary human macrophages (27). Therefore, inhibition of the release of different proinflammatory cytokines may be associated with activation of TGR5 by various agonists in human monocytes/macrophages, with or without RA. The present findings indicated that TGR5 is a potential suppressor of NF-κB-dependent inflammatory responses; however, the nature of the interaction between TGR5 and NF-κB/IκBα in the PBMCs of patients with RA remains unknown. It has been reported that TGR5 serves an important role in immunological regulation via cyclic adenosine monophosphate, a second messenger with immunoregulatory functions (25,43). Additionally, a previous study indicated that TGR5 promotes the activation of cytosolic protein β-arrestin 2, which interacts with IκBα following bacterial antigen stimulation in mouse livers, suppressing the production of TNF-α (26). Therefore, further studies into the mechanisms underlying the modulation of the activity of NF-κB and IκBα in the PBMCs of patients with RA by TGR5 are required.

NF-κB is a family of ubiquitously expressed transcription factors that serve crucial roles in the majority of inflammatory and immune responses (15). Constitutive NF-κB activation has been associated with the pathogenesis of numerous diseases, including inflammatory and rheumatic diseases (RA and osteoarthritis), atherosclerosis, asthma, multiple sclerosis, chronic inflammatory demyelinating inflammatory bowel disease and diabetes (14). Additionally, studies on emerging animal models of inflammatory arthritis support the hypothesis that NF-κB is a predominant adaptor in the development and progression of arthritis in vivo (44,45). In the present study, it was revealed that TGR5 activation significantly inhibited NF-κB activity in vitro, implying that TGR5 activation may potentially alleviate arthritis symptoms and inflammation of RA in vivo. CIA mouse models with pathological features similar to those of human RA are used to study arthritic diseases. It was observed that TGR5 activation by LCA significantly suppressed inflammation and the progression of arthritis in mice with CIA, as determined by their arthritis score. RA is characterized by chronic inflammatory synovitis and joint destruction; thus, future experiments will aim to identify the mechanisms underlying the effects of TGR5 on synovitis and joint destruction in patients with RA or mice with CIA.

In RA, activated immune cells secrete large quantities of proinflammatory cytokines. TNF-α is a potent inducer of inflammation, and IL-6 and IL-1β are downstream mediators of TNF-α, together serving significant roles in the progression of inflammatory synovitis and joint destruction in RA (46,47). IL-8 is involved in leukocyte recruitment to a diseased synovium (48). Agents that inhibit TNF-α, IL-6 and IL-1 are frequently used in clinical settings therapeutically (49). In the present study, TGR5 activation demonstrated therapeutic effects against RA by downregulating the levels of TNF-α, IL-1β, IL-6 and IL-8 in the plasma of mice with CIA in vivo. TGR5 activation also significantly suppressed LPS-induced TNF-α, IL-1β, IL-6 and IL-8 expression in PBMCs in vitro. These results indicated that TGR5 activation may inhibit proinflammatory cytokine expression in monocytes/macrophages and further reduce inflammation in RA; however, the influence of TGR5 on other immune cells and tissues of patients with RA or mice with CIA remains unclear and requires further study. Monocytes can differentiate into proinflammatory microbicidal M1 macrophages or anti-inflammatory M2 macrophage subtypes (50). Macrophages are stimulated to differentiate into M1 macrophages by infectious microorganism-associated molecules such as LPS and inflammatory cytokines such as interferon-γ (51). M1 macrophages can produce inflammatory cytokines, including interleukin IL-1β, TNF-a and IL-6 (52). Högenauer et al (42) demonstrated that TGR5 activation stabilizes the phenotype of noninflammatory M2-like type cells during the differentiation of monocytes into macrophages. The effect of TGR5 on macrophage differentiation in mice with CIA requires further investigation.

Collectively, TGR5 activation attenuated the expression of TNF-α, IL-1β, IL-6 and IL-8 via inhibition of NF-κB activity in LPS-stimulated PBMCs in vitro, decreased the levels of TNF-α, IL-1β, IL-6 and IL-8 in the plasma of mice with CIA in vivo, and hindered the progression of arthritis in mouse models of CIA, as determined by the arthritis score. RA has also been reported to have improved during jaundice, suggesting that TGR5 activation may provide a novel target for the treatment of RA (5355). Future studies should investigate cases of RA with obstructive jaundice, to further determine the potential role and effects of TGR5 in RA.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the National Natural Science Foundation of China (grant no. 81401631) and Education of Zhejiang Province (grant no. Y201737942).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JJZ and ZYL participated in study development and collection of patient samples, performed experiments, and actively wrote the first draft of the manuscript. CLL was involved in collecting and analyzing part of the clinical data. LMZ participated in analyzing the data from in vitro cell experiments and revising the manuscript. All authors approved the final manuscript.

Ethics approval and consent to participate

The study protocol was approved by the the Institutional Animal and Clinical Committees of Hunan Cancer Hospital and written informed consent was obtained from all participants in the study.

Patient consent for publication

Written informed consent was obtained from all participants.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

RA

rheumatoid arthritis

PBMC

peripheral blood mononuclear cell

HC

healthy control

TGR5

G-protein-coupled bile acid receptor 1

RF

rheumatoid factor

CRP

C-reactive protein

ESR

erythrocyte sedimentation rate

MMP

matrix metalloproteinase

anti-CCP

anti-cyclic citrullinated peptide

DAS28

28-Joint Disease Activity Score

ELISA

enzyme-linked immunosorbent assay

HAQ

Health Assessment Questionnaire

RT-qPCR

reverse transcription-quantitative polymerase chain reaction

NF-κB

nuclear factor-κB

TNF-α

tumor necrosis factor-α

IL

interleukin

IκBα

IκB kinase α

CIA

collagen II-induced arthritis

ACR

American College of Rheumatology

28TJC

28-joint tender joint count

28SJC

28-joint swollen joint count

PtGA

patient global assessment of disease activity

PrGA

provider global assessment of disease activity

cDNA

complementary DNA

CII

collagen type II

DMARD

disease-modifying anti-rheumatic drug

FLS

fibroblast-like synoviocyte

LCA

lithocholic acid

LPS

lipopolysaccharide

References

1 

Siebert S, Tsoukas A, Robertson J and McInnes I: Cytokines as therapeutic targets in rheumatoid arthritis and other inflammatory diseases. Pharmacol Rev. 67:280–309. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Choy EH and Panayi GS: Cytokine pathways and joint inflammation in rheumatoid arthritis. N Engl J Med. 344:907–916. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Karsdal MA, Woodworth T, Henriksen K, Maksymowych WP, Genant H, Vergnaud P, Christiansen C, Schubert T, Qvist P, Schett G, et al: Biochemical markers of ongoing joint damage in rheumatoid arthritis-current and future applications, limitations and opportunities. Arthritis Res Ther. 13:2152011. View Article : Google Scholar : PubMed/NCBI

4 

Wang Q, Ma Y, Liu D, Zhang L and Wei W: The roles of B cells and their interactions with fibroblast-like synoviocytes in the pathogenesis of rheumatoid arthritis. Int Arch Allergy Immunol. 155:205–211. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Lefevre S, Meier FM, Neumann E and Muller-Ladner U: Role of synovial fibroblasts in rheumatoid arthritis. Curr Pharm Des. 21:130–141. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Roberts CA, Dickinson AK and Taams LS: The interplay between monocytes/macrophages and CD4(+) T cell subsets in rheumatoid arthritis. Front Immunol. 6:5712015. View Article : Google Scholar : PubMed/NCBI

7 

Davignon JL, Hayder M, Baron M, Boyer JF, Constantin A, Apparailly F, Poupot R and Cantagrel A: Targeting monocytes/macrophages in the treatment of rheumatoid arthritis. Rheumatology (Oxford). 52:590–598. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Kinne RW, Stuhlmuller B and Burmester GR: Cells of the synovium in rheumatoid arthritis. Macrophages. Arthritis Res Ther. 9:2242007. View Article : Google Scholar : PubMed/NCBI

9 

Schett G: Cells of the synovium in rheumatoid arthritis. Osteoclasts. Arthritis Res Ther. 9:2032007. View Article : Google Scholar : PubMed/NCBI

10 

Yokota K: Inflammation and osteoclasts. Nihon Rinsho Men'eki Gakkai kaishi. 40:367–376. 2017.(In Japanese). View Article : Google Scholar

11 

Alam J, Jantan I and Bukhari SNA: Rheumatoid arthritis: Recent advances on its etiology, role of cytokines and pharmacotherapy. Biomed Pharmacother. 92:615–633. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Mulherin D, Fitzgerald O and Bresnihan B: Synovial tissue macrophage populations and articular damage in rheumatoid arthritis. Arthritis Rheum. 39:115–124. 1996. View Article : Google Scholar : PubMed/NCBI

13 

Yanni G, Whelan A, Feighery C and Bresnihan B: Synovial tissue macrophages and joint erosion in rheumatoid arthritis. Ann Rheum Dis. 53:39–44. 1994. View Article : Google Scholar : PubMed/NCBI

14 

Aravilli RK, Vikram SL and Kohila V: Phytochemicals as potential antidotes for targeting NF-κB in rheumatoid arthritis. 3 Biotech. 7:2532017. View Article : Google Scholar : PubMed/NCBI

15 

Roman-Blas JA and Jimenez SA: NF-κB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis and Cartilage. 14:839–848. 2006. View Article : Google Scholar : PubMed/NCBI

16 

McInnes IB and Schett G: Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol. 7:429–442. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Schaap FG, Trauner M and Jansen PL: Bile acid receptors as targets for drug development. Nat Rev Gastroenterol Hepatol. 11:55–67. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Lin TH, Tang CH, Wu K, Fong YC, Yang RS and Fu WM: 15-deoxy-Δ(12,14)-prostaglandin-J2 and ciglitazone inhibit TNF-α-induced matrix metalloproteinase 13 production via the antagonism of NF-κB activation in human synovial fibroblasts. J Cell Physiol. 226:3242–3250. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Yu DD, Sousa KM, Mattern DL, Wagner J, Fu X, Vaidehi N, Forman BM and Huang W: Stereoselective synthesis, biological evaluation, and modeling of novel bile acid-derived G-protein coupled bile acid receptor 1 (GP-BAR1, TGR5) agonists. Bioorg Med Chem. 23:1613–1628. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Pols TW: TGR5 in inflammation and cardiovascular disease. Biochem Soc Trans. 42:244–249. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Russell DW and Setchell KD: Bile acid biosynthesis. Biochemistry. 31:4737–4749. 1992. View Article : Google Scholar : PubMed/NCBI

22 

Pols TW, Noriega LG, Nomura M, Auwerx J and Schoonjans K: The bile acid membrane receptor TGR5 as an emerging target in metabolism and inflammation. J Hepatol. 54:1263–1272. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Kawamata Y, Fujii R, Hosoya M, Harada M, Yoshida H, Miwa M, Fukusumi S, Habata Y, Itoh T, Shintani Y, et al: A G protein-coupled receptor responsive to bile acids. J Biol Chem. 278:9435–9440. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Keitel V, Donner M, Winandy S, Kubitz R and Haussinger D: Expression and function of the bile acid receptor TGR5 in Kupffer cells. Biochem Biophys Res Commun. 372:78–84. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Pols TW, Nomura M, Harach T, Lo Sasso G, Oosterveer MH, Thomas C, Rizzo G, Gioiello A, Adorini L, Pellicciari R, et al: TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. 14:747–757. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Wang YD, Chen WD, Yu D, Forman BM and Huang W: The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor κ light-chain enhancer of activated B cells (NF-κB) in mice. Hepatology. 54:1421–1432. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Haselow K, Bode JG, Wammers M, Ehlting C, Keitel V, Kleinebrecht L, Schupp AK, Häussinger D and Graf D: Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. J Leukoc Boil. 94:1253–1264. 2013. View Article : Google Scholar

28 

Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH and Luthra HS: The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 31:315–324. 1988. View Article : Google Scholar : PubMed/NCBI

29 

Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO III, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, et al: 2010 Rheumatoid arthritis classification criteria: An American college of rheumatology/european league against rheumatism collaborative initiative. Arthritis Rheum. 62:2569–2581. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Koh ET, Seow A, Pong LY, Koh WH, Chan L, Howe HS, Lim TH and Low CK: Cross cultural adaptation and validation of the Chinese health assessment questionnaire for use in rheumatoid arthritis. J Rheumatol. 25:1705–1708. 1998.PubMed/NCBI

31 

Anderson J, Caplan L, Yazdany J, Robbins ML, Neogi T, Michaud K, Saag KG, O'Dell JR and Kazi S: Rheumatoid arthritis disease activity measures: American college of rheumatology recommendations for use in clinical practice. Arthritis Care Res (Hoboken). 64:640–647. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Jones-Bolin S: Guidelines for the care and use of laboratory animals in biomedical research. Curr Protoc Pharmacol Appendix 4: Appendix 4B. 2012. View Article : Google Scholar

34 

Brand DD, Latham KA and Rosloniec EF: Collagen-induced arthritis. Nat Protoc. 2:1269–1275. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Yoo SA, Park BH, Park GS, Koh HS, Lee MS, Ryu SH, Miyazawa K, Park SH, Cho CS and Kim WU: Calcineurin is expressed and plays a critical role in inflammatory arthritis. J Immunol. 177:2681–2690. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Lee H, Nah SS, Chang SH, Kim HK, Kwon JT, Lee S, Cho IH, Lee SW, Kim YO, Hong SJ and Kim HJ: PER2 is downregulated by the LPS-induced inflammatory response in synoviocytes in rheumatoid arthritis and is implicated in disease susceptibility. Mol Med Rep. 16:422–428. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Cho YG, Cho ML, Min SY and Kim HY: Type II collagen autoimmunity in a mouse model of human rheumatoid arthritis. Autoimmun Rev. 7:65–70. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Mo YQ, Dai L, Zheng DH, Zhu LJ, Wei XN, Pessler F, Shen J and Zhang BY: Synovial infiltration with CD79a-positive B cells, but not other B cell lineage markers, correlates with joint destruction in rheumatoid arthritis. J Rheumatol. 38:2301–2308. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Zhu LJ, Dai L, Zheng DH, Mo YQ, Ou-Yang X, Wei XN, Shen J and Zhang BY: Upregulation of tumor necrosis factor receptor-associated factor 6 correlated with synovitis severity in rheumatoid arthritis. Arthritis Res Ther. 14:R1332012. View Article : Google Scholar : PubMed/NCBI

40 

Smeets TJ, Barg EC, Kraan MC, Smith MD, Breedveld FC and Tak PP: Analysis of the cell infiltrate and expression of proinflammatory cytokines and matrix metalloproteinases in arthroscopic synovial biopsies: Comparison with synovial samples from patients with end stage, destructive rheumatoid arthritis. Ann Rheum Dis. 62:635–638. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Proudman SM, Cleland LG and Mayrhofer G: Effects of tumor necrosis factor-alpha, interleukin 1beta, and activated peripheral blood mononuclear cells on the expression of adhesion molecules and recruitment of leukocytes in rheumatoid synovial xenografts in SCID mice. J Rheumatol. 26:1877–1889. 1999.PubMed/NCBI

42 

Högenauer K, Arista L, Schmiedeberg N, Werner G, Jaksche H, Bouhelal R, Nguyen DG, Bhat BG, Raad L, Rauld C and Carballido JM: G-protein-coupled bile acid receptor 1 (GPBAR1, TGR5) agonists reduce the production of proinflammatory cytokines and stabilize the alternative macrophage phenotype. J Med Chem. 57:10343–10354. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Ichikawa R, Takayama T, Yoneno K, Kamada N, Kitazume MT, Higuchi H, Matsuoka K, Watanabe M, Itoh H, Kanai T, et al: Bile acids induce monocyte differentiation toward interleukin-12 hypo-producing dendritic cells via a TGR5-dependent pathway. Immunology. 136:153–162. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Makarov SS: NF-kappa B in rheumatoid arthritis: A pivotal regulator of inflammation, hyperplasia, and tissue destruction. Arthritis Res. 3:200–206. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Mor A, Abramson SB and Pillinger MH: The fibroblast-like synovial cell in rheumatoid arthritis: A key player in inflammation and joint destruction. Clin Immunol. 115:118–128. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Zheng Y, Sun L, Jiang T, Zhang D, He D and Nie H: TNFalpha promotes Th17 cell differentiation through IL-6 and IL-1beta produced by monocytes in rheumatoid arthritis. J Immunol Res. 2014:3853522014. View Article : Google Scholar : PubMed/NCBI

47 

Xin W, Huang C, Zhang X, Xin S, Zhou Y, Ma X, Zhang D, Li Y, Zhou S, Zhang D, et al: Methyl salicylate lactoside inhibits inflammatory response of fibroblast-like synoviocytes and joint destruction in collagen-induced arthritis in mice. Br J Pharmacol. 171:3526–3538. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Szekanecz Z, Kim J and Koch AE: Chemokines and chemokine receptors in rheumatoid arthritis. Semin Immunol. 15:15–21. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Smolen JS, Landewe R, Bijlsma J, Burmester G, Chatzidionysiou K, Dougados M, Nam J, Ramiro S, Voshaar M, van Vollenhoven R, et al: EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 76:960–977. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Italiani P and Boraschi D: From monocytes to M1/M2 macrophages: Phenotypical vs. Functional differentiation. Front Immunol. 5:5142014. View Article : Google Scholar : PubMed/NCBI

51 

Dey A, Allen J and Hankey-Giblin PA: Ontogeny and polarization of macrophages in inflammation: Blood monocytes versus tissue macrophages. Front Immunol. 5:6832015. View Article : Google Scholar : PubMed/NCBI

52 

Gordon S and Taylor PR: Monocyte and macrophage heterogeneity. Nat Rev Immunol. 5:953–964. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Crocker I, Lawson N and Fletcher J: Effect of pregnancy and obstructive jaundice on inflammatory diseases: The work of P S hench revisited. Ann Rheum Dis. 61:307–310. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Rutkauskaite E, Zacharias W, Schedel J, Müller-Ladner U, Mawrin C, Seemayer CA, Alexander D, Gay RE, Aicher WK, Michel BA, et al: Ribozymes that inhibit the production of matrix metalloproteinase 1 reduce the invasiveness of rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 50:1448–1456. 2004. View Article : Google Scholar : PubMed/NCBI

55 

Ma JD, Zhou JJ, Zheng DH, Chen LF, Mo YQ, Wei XN, Yang LJ and Dai L: Serum matrix metalloproteinase-3 as a noninvasive biomarker of histological synovitis for diagnosis of rheumatoid arthritis. Mediators Inflammation. 2014:1792842014. View Article : Google Scholar

Related Articles

Journal Cover

November-2019
Volume 20 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li ZY, Zhou JJ, Luo CL and Zhang LM: Activation of TGR5 alleviates inflammation in rheumatoid arthritis peripheral blood mononuclear cells and in mice with collagen II‑induced arthritis. Mol Med Rep 20: 4540-4550, 2019
APA
Li, Z., Zhou, J., Luo, C., & Zhang, L. (2019). Activation of TGR5 alleviates inflammation in rheumatoid arthritis peripheral blood mononuclear cells and in mice with collagen II‑induced arthritis. Molecular Medicine Reports, 20, 4540-4550. https://doi.org/10.3892/mmr.2019.10711
MLA
Li, Z., Zhou, J., Luo, C., Zhang, L."Activation of TGR5 alleviates inflammation in rheumatoid arthritis peripheral blood mononuclear cells and in mice with collagen II‑induced arthritis". Molecular Medicine Reports 20.5 (2019): 4540-4550.
Chicago
Li, Z., Zhou, J., Luo, C., Zhang, L."Activation of TGR5 alleviates inflammation in rheumatoid arthritis peripheral blood mononuclear cells and in mice with collagen II‑induced arthritis". Molecular Medicine Reports 20, no. 5 (2019): 4540-4550. https://doi.org/10.3892/mmr.2019.10711