ORIGINAL ARTICLE

Antcin K inhibits VCAM-1-dependent monocyte adhesion in human rheumatoid arthritis synovial fibroblasts

David Achudhan1, Sunny Li-Yun Chang1, Shan-Chi Liu2, Yen-You Lin3, Wei-Chien Huang1, Yang-Chang Wu1,4,5, Chien-Chung Huang3,6, Chun-Hao Tsai7,8, Chih-Yuan Ko8, Yueh-Hsiung Kuo5,9,10,11* and Chih-Hsin Tang1,3,5,11*

1Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung, Taiwan; 2Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan; 3School of Medicine, China Medical University, Taichung, Taiwan; 4Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan; 5Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; 6Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; 7Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan; 8Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan; 9Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; 10Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan; 11Chinese Medicine Research Center, China Medical University, Taichung, Taiwan

Popular scientific summary

Abstract

Background: Antcin K, an extract of Antrodia cinnamomea (a medicinal mushroom endemic to Taiwan commonly used in Chinese medicine preparations), inhibits proinflammatory cytokine production and angiogenesis in human rheumatoid arthritis synovial fibroblasts (RASFs), major players in RA disease. Antcin K also inhibits disease activity in mice with collagen-induced arthritis (CIA). Up until now, the effects of Antcin K upon cell adhesion molecules (CAMs) were unknown.

Methods: RA and healthy synovial tissue samples (n = 10 in each group) were retrieved from the Gene Expression Omnibus (GEO) database (accession code: GDS5401) to compare CAM and monocyte marker expressions. In addition, synovial tissue samples from six RA patients and six patients undergoing arthroscopy for trauma/joint derangement (healthy controls) were subjected to immunohistochemical (IHC) analysis. mRNA and protein expression levels were analyzed in RASFs using RT-qPCR (Reverse transcription-quantitative polymerase chain reaction) and Western blot. RASFs were incubated with Antcin K and examined for monocyte adherence by fluorescence microscopy. Ankle joint tissue specimens from a CIA mouse model and healthy controls were stained with hematoxylin and eosin (H&E) and Safranin-O/Fast Green to examine histological changes and evidence of bone loss. IHC analysis determined levels of vascular cell adhesion molecule 1 (VCAM-1) and CD11b in CIA ankle tissue and clinical synovial tissue.

Results: Levels of VCAM-1 expression were higher in the GEO database specimens and the study’s clinical samples of RA synovial tissue compared with the healthy specimens. Antcin K dose-dependently inhibited VCAM-1 expression and monocyte adhesion in RASFs. Antcin K also significantly inhibited levels of VCAM-1 and monocyte CD11b expression in CIA tissue. These effects appeared to be mediated by MEK1/2-ERK, p38, and AP-1 signaling.

Conclusions: Antcin K seems promising for the treatment of RA and deserves further investigations.

Keywords: rheumatoid arthritis; cell adhesion molecules; monocytes; VCAM-1; CD11b; Antrodia cinnamomea; Antcin K

 

Citation: Food & Nutrition Research 2022, 66: 8645 - http://dx.doi.org/10.29219/fnr.v66.8645

Copyright: © 2022 David Achudhan et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material for any purpose, even commercially, provided the original work is properly cited and states its license.

Received: 4 March 2022; Revised: 14 April 2022; Accepted: 14 April 2022; Published: 2 June 2022

Competing interests and funding: The authors declare no conflict of interest. This research was funded by Ministry of Science and Technology in Taiwan under grant numbers MOST 110-2320-B-039-022-MY3, MOST 110-2314-B-039-008, and MOST 110-2314-B-039-012; the DMR-111-165; and DMR-111-117; DMR-111-028 was funded by China Medical University Hospital and from China Medical University under the Higher Education Sprout Project, Ministry of Education, Taiwan (CMRC-CHM-2-2).

*Yueh-Hsiung Kuo, Institute: Tsuzuki Institute for Traditional Medicine, Address: 91, Hsueh-Shih Road, Taichung,404, Taiwan, Republic of China. Email: kuoyh@mail.cmu.edu.tw. Tel: 886-4-22053366 ext 5701, 5709

Chih-Hsin Tang, Institute: Department of Pharmacology, School of Medicine, China Medical University, Address: #91, Hsueh-Shih Road, Taichung city 40402, Taiwan. E-mail: chtang@mail.cmu.edu.tw. Tel: +886-4-22053366#7726.

These authors contributed equally to this work.

To access the supplementary material, please visit the article landing page

 

Rheumatoid arthritis (RA) is a degenerative joint disorder that affects an estimated 1% of people worldwide (1, 2). RA is characterized by synovial hyperplasia, joint damage, cartilage degradation, and bone erosion with infiltration of monocytes into the synovium (3, 4). The activation of RA synovial fibroblasts (RASFs) by inflammatory factors subsequently activates and increases the expression of cell adhesion molecules (CAMs), including, integrins and vascular cell adhesion molecule 1 (VCAM-1), which mediate adhesion to the extracellular matrix (ECM) in the inflamed RA synovium and facilitate joint damage (5). Higher serum levels of VCAM-1 have been found in patients with RA than in patients without RA; moreover, serum VCAM-1 levels are significantly and positively correlated with RA severity, and the levels gradually decrease with treatment (6, 7). VCAM‑1 expression is upregulated in RASFs activated by proinflammatory cytokines including interleukin (IL)‑4, tumor necrosis factor (TNF)‑α, TNF‑β, and other proinflammatory cytokines (5, 8). Disease-modifying antirheumatic drugs (DMARDs) are known to markedly lower VCAM-1 levels in patients with RA (9), while the humanized monoclonal antibody adalimumab significantly prevents the upregulation of VCAM-1 mRNA and protein expression after TNF-α stimulation of human umbilical vein endothelial cells (HUVECs) (10). However, these RA treatments are also associated with undesirable adverse effects, including gastrointestinal problems (nausea, diarrhea, and abdominal pain), rash and allergic reactions, alopecia, suppression of bone marrow, liver cirrhosis, and hepatotoxicity, as well as a higher incidence of common and sometimes serious infections (11); these adverse drug reactions can result in treatment discontinuation (12). Novel anti-arthritic drugs without adverse effects and good tolerability are needed to heal RA patients.

Traditional Chinese medicine (TCM) contains functional compounds that can effectively treat RA (13), with evidence of attenuation of disease activity and protection against bone degradation (14, 15). Notably, our previous research has found that an extract from Antrodia cinnamomea (syn. Antrodia camphorata, a medicinal mushroom endemic to Taiwan commonly used in TCM preparations), Antcin K, has demonstrated anti-inflammatory effects in human RASFs and in collagen-induced arthritis (CIA) mice by inhibiting proinflammatory cytokine expression (16) and also anti-angiogenic effects by inhibiting vascular endothelial growth factor (VEGF) expression in RASFs (17). These results imply that Antcin K achieves these effects by inhibiting CAMs and monocyte expression, but no research has yet investigated Antcin K and its underlying mechanisms in RA.

Methods

Antibodies, chemicals, and reagents

Antibodies used in this study are listed in Supplementary Table 1. All pharmacological activators and 2´,7´-Bis(2-carboxyethyl)-5(6)-carboxyfluorescein tetrakis(acetoxymethyl) ester (BCECF-AM) were obtained from Sigma-Aldrich (St. Louis, MO, USA). The TRIzol kit was obtained from MDBio Inc. (Taipei, Taiwan). Enhanced chemiluminescence (ECL) reagents were obtained from Merck Millipore (WBKLS0500, Billerica, MA, USA). A Novolink Polymer Detection System Kit was supplied by Leica Microsystems. Fetal bovine serum (FBS) and other components of culture media were purchased from Gibco and Thermo Fisher Scientific, Inc. (Waltham, MA, USA).

Cell culture

The human RASF cell line MH7A was purchased from Riken (Ibaraki, Japan) and cultured in RPMI-1640 medium supplemented with 10% FBS and penicillin/streptomycin 100 U/mL (16, 18). The human monocyte cell line THP-1 was obtained from the American Type Culture Collection (Manassas, VA, USA) and augmented with RPMI-1640 medium containing 10% FBS. Both cell lines were incubated for 24 h at 37°C in a humidified atmosphere with 5% CO2 (1921).

Analysis of the Gene Expression Omnibus dataset

We downloaded RA and healthy clinical synovial tissue data from the public Gene Expression Omnibus (GEO) dataset (accession code: GDS5401) to compare CAM and monocyte marker expression between the RA and healthy samples (n = 10 for both samples) (22, 23).

Clinical samples

Clinical samples were obtained from six RA patients and from six patients undergoing arthroscopy for trauma/joint derangement (who served as healthy controls) in China Medical University Hospital, Taichung, Taiwan.

mRNA expression analysis

The TRIzol kit extracted total RNA from RASFs, and then 1 μg of total RNA was reverse-transcribed into double-stranded (ds) cDNA using an oligo (dT) primer. The RT-qPCR (Reverse transcription-quantitative polymerase chain reaction) assay was performed according to our previous reports (19, 24). The primers employed in the RT-qPCR are listed in Supplementary Table 2.

Protein expression analysis

Total proteins were extracted by the RIPA Lysis Buffer, and then the proteins were resolved and separated by SDS-PAGE. Protein expression was examined by Western blot, according to our published methods (25, 26). Details of antibody sources and dilution factors are listed in Supplementary Table 1.

Cell adhesion assay

RASFs were incubated with various concentrations of Antcin K for 24 h at 37°C and then with BCECF-AM-labeled THP-1 cells for 1 h at 37°C. Non-adherent cells were removed by PBS washing. Attached THP-1 cells in RASFs were counted using a Nikon Eclipse Ti-S Fluorescent microscope (Nikon, Japan) with images captured at 10× magnification. Captured images were counted and quantified by MacBiophotonics ImageJ software (v1.51, National Institutes of Health, Bethesda, MD, USA) (19, 20, 27).

Immunohistochemical analysis

Ankle joint tissue was stained with hematoxylin and eosin (H&E) and Safranin-O/Fast Green for histological changes and evidence of bone loss. The tissue was subjected to immunohistochemical (IHC) analysis with VCAM-1 and CD11b antibodies, according to previously described methodology (28, 29).

Statistical analysis

All expressed values are presented as mean ± standard deviation (SD). The Student’s t-test was used to compare the means between the study groups. The statistical difference was considered significant when the P-value was <0.05.

Results

High levels of VCAM-1 and CD11b expressions in RA synovial tissue

Higher serum levels of VCAM-1 reflect increasingly severe RA disease and its characteristic synovial inflammation (6, 30). We, therefore, screened for protein expression markers of the four main CAM classes (cadherins, integrins, selectins, and CAMs of the immunoglobulin superfamily) in records downloaded from the GEO database (accession code: GDS5401). The data revealed significantly higher levels of VCAM-1 expression in RA synovial tissue than in samples from healthy controls (Fig. 1a, b) as well as significantly higher levels of the monocyte marker CD11b in RA tissue compared with healthy tissue (Fig. 1a, c). Similarly, in our IHC staining of synovial tissue from RA patients and healthy controls, levels of VCAM-1 and CD11b were significantly higher in the RA tissue than in the healthy tissue (Fig. 2a–d).


Fig 1
Fig. 1. Levels of cell adhesion molecule expression in RA synovial tissue. (a) Records downloaded from the GEO database (accession code: GDS5401) were screened for protein expression markers of the four main CAM classes (cadherins, integrins, selectins, and CAMs of the immunoglobulin superfamily) in synovial tissue samples from 10 patients with RA and 10 healthy individuals (b, c). Levels of VCAM-1 and CD11b expressions in RA and normal synovial tissues. RA, rheumatoid arthritis; NL, normal controls. *P < 0.05 compared with normal controls.

Fig 2
Fig. 2. Upregulation of VCAM-1 and CD11b expressions in human RA synovial tissue. (a) Immunostaining of VCAM-1 expression in human RA (n = 6) and normal tissue (n = 6). Black arrows indicate levels of VCAM-1 expression in the synovial tissue. (b) Quantification of VCAM-1 expression by IHC score. (c) IHC staining of CD11b expression in RA (n = 6) and normal tissue (n = 6). Black arrows indicate levels of CD11b expression in the synovial tissue. (d) Quantification of CD11b expression by IHC score. RA, rheumatoid arthritis; NL, normal controls. *P < 0.05 compared with normal controls.

Antcin K inhibits VCAM-1 expression and monocyte adhesion in human RASFs

Soluble VCAM-1 is responsible for a large proportion of monocyte chemotaxis in RASFs (31). This study examined the relationship between drug exposure (Antcin K at concentrations of 0, 0.3, 1, 3, or 10 μM) and pharmacologic or toxicologic responses. The 10 μM concentration of Antcin K was selected for evaluation alone and in combination with pathway activators, based on the results of our previous studies, showing that Antcin K (10 μM) effectively inhibited pro-inflammatory cytokine expressions without affecting RASF cell viability (16, 17). Antcin K significantly suppressed VCAM-1 mRNA and protein levels in a dose-dependent manner (Fig. 3a, b) and also significantly inhibited the adhesion of THP-1 monocytic cells to RASFs (Fig. 3c, d). Thus, Antcin K appears to inhibit VCAM-1 expression and VCAM-1-mediated monocyte adhesion to human RASFs.

Fig 3
Fig. 3. Antcin K inhibits VCAM-1 expression and monocyte adhesion in human RASFs. (a, b) Antcin K (0.3–10 μM) was administered to RASFs for 24 h, and then VCAM-1 expression was examined using RT-qPCR (n = 4) and Western blot analyses (n = 3). (c, d) RASFs were incubated with indicated concentrations of Antcin K for 24 h. BCECF-AM-labeled THP-1 cells were then added to the RASFs for 1 h. Monocyte adherence in cultured RASFs was examined by fluorescence microscopy and photographed (n = 4). *P < 0.05 compared with the control.

Antcin K inhibits VCAM-1 synthesis by suppressing MEK1/2-ERK and p38 signaling

The Mitogen-activated protein kinase kinase (MEK)/Extracellular signal–regulated kinase (ERK) signaling cascades are important for inflammatory progression in RA (32, 33). We, therefore, sought to determine whether Antcin K affects MEK and ERK signaling. Incubation of RASFs with Antcin K inhibited MEK1/2 and ERK phosphorylation (Figs. 4a and 5a). We then stimulated the RASFs for 30 min with the MEK activator PAF C-16 (10 μM) and the ERK activator ceramide C6 (10 μM); both significantly antagonized Antcin K-induced reductions in VCAM-1 expression (Figs. 4b–e and 5b–e). The p38 signaling pathway is involved in the regulation of CAM and proinflammatory cytokine expression during RA development (33, 34). We, therefore, investigated whether Antcin K inhibits VCAM-1 expression and monocyte adhesion in RASFs via the p38 signaling pathway. We incubated RASFs with varying concentrations of Antcin K and found that Antcin K dose-dependently inhibited p38 phosphorylation (Fig. 6a). Pretreatment with the p38 activator anisomycin reversed Antcin K-induced inhibition of VCAM-1 expression and adhesion of monocytes in RASFs (Fig. 6b–e). These findings suggest that Antcin K inhibits VCAM-1 and monocyte adhesion in human RASFs via the MEK1/2-ERK and p38 signaling pathways.

Fig 4
Fig. 4. The MEK1/2 pathway is involved in Antcin K-induced VCAM-1 inhibition. (a) Antcin K (0.3–10 μM) was administered to RASFs, and MEK1/2 phosphorylation was analyzed by Western blot analysis (n = 3). (b–e) RASFs were stimulated with the MEK activator (PAF C-16) for 30 min and then incubated with 10 μM of Antcin K for 24 h. VCAM-1 levels were analyzed by RT-qPCR (n = 4) and Western blot (n = 3). Monocyte adhesion in cultured RASFs was determined by the monocyte adhesion assay (n = 4). *P < 0.05 compared with the control; # P < 0.05 compared with Antcin K treatment.

Fig 5
Fig. 5. The ERK pathway is involved in Antcin K-induced VCAM-1 inhibition. (a) RASFs were treated with Antcin K (0.3–10 μM) for 24 h, and then ERK phosphorylation was examined by Western blot (n = 3). (b–e) RASFs were stimulated with the ERK activator (ceramide C6) for 30 min and then incubated with 10 μM of Antcin K for 24 h. VCAM-1 levels were analyzed by RT-qPCR (n = 4) and Western blot (n = 3). Monocyte adhesion in cultured RASFs was determined by the monocyte adhesion assay (n = 4). *P < 0.05 compared with the control; # P < 0.05 compared with Antcin K treatment.

Fig 6
Fig. 6. The p38 pathway is involved in Antcin K-induced VCAM-1 inhibition. (a) RASFs were treated with Antcin K (0.3–10 μM) for 24 h, and then p38 phosphorylation was examined by Western blot (n = 3). (b–e) RASFs were stimulated with the p38 activator (anisomycin) for 30 min and then incubated with Antcin K (10 μM) for 24 h. VCAM-1 levels were examined by RT-qPCR (n = 4) and Western blot (n = 3). Monocyte adhesion in cultured RASFs was examined by the monocyte adhesion assay (n = 4). *P < 0.05 compared with the control; # P < 0.05 compared with Antcin K treatment.

Antcin K inhibits VCAM-1 synthesis by suppressing AP-1 signaling

AP-1 is a critical transcription factor that regulates the production of CAMs and proinflammatory cytokines during RA progression (35, 36). We, therefore, sought to determine whether AP-1 controls the effects of Antcin K-induced VCAM-1 expression. Incubation of RASFs with Antcin K dose-dependently inhibited c-Jun phosphorylation (Fig. 7a). After stimulating the RASFs with an AP-1 activator (D-erythro-SPC) for 30 min, treatment with Antcin K downregulated VCAM-1 expression and monocyte adhesion in the RASFs (Fig. 7b–e). To confirm the involvement of the AP-1 pathway, we transfected the RASFs with the AP-1 plasmid prior to checking luciferase activity. Antcin K dose-dependently inhibited AP-1 luciferase activity (Fig. 7f). Thus, Antcin K-induced decreases in VCAM-1 expression and monocyte adhesion to RASFs appear to occur through the MEK1/2-ERK, p38, and AP-1 signaling pathways.

Fig 7
Fig. 7. The AP-1 pathway is involved in Antcin K-induced VCAM-1 inhibition. (a) RASFs were treated with Antcin K (0.3–10 μM) for 24 h, and then c-Jun phosphorylation was examined by Western blot (n = 3). (b–e) RASFs were stimulated with the AP-1 activator (D-erythro-SPC) for 30 min and then incubated with Antcin K (10 μM) for 24 h. VCAM-1 levels were examined by RT-qPCR (n = 4) and Western blot (n = 3). Monocyte adhesion in cultured RASFs was examined by the monocyte adhesion assay (n = 4). (f) RASFs were transfected with the AP-1 luciferase plasmid and then treated with Antcin K at the indicated concentrations, and luciferase activity was quantified. *P < 0.05 compared with the control; # P < 0.05 compared with Antcin K treatment.

Antcin K attenuates VCAM-1 and CD11b expressions in CIA mice

We have previously demonstrated that at doses of 10 mg/kg and 30 mg/kg, Antcin K significantly attenuates the signs and symptoms of RA (16). In this study, we examined VCAM-1 and CD11b expressions in ankle joint tissue samples obtained from the study groups in our previous research (controls, untreated CIA mice, and CIA mice treated with Antcin K 10 or 30 mg/kg). H&E staining and Safranin-O staining revealed that both dosages of Antcin K reversed the bone volume loss exhibited by the control CIA group (Fig. 8a). Similarly, IHC findings showed that Antcin K inhibited VCAM-1 and CD11b expressions in ankle joint tissue as compared with untreated CIA mice, in a dose-dependent manner (Fig. 8b–c). These findings suggest that the antiarthritic effects of Antcin K are related to its inhibition of VCAM-1 expression and monocyte infiltration into the synovium.

Fig 8
Fig. 8. Antcin K reduces histological severity of RA disease. (a) Specimens from control ankle joints, CIA ankle joints, and Antcin K-treated CIA ankle joints were counterstained with H&E and Safranin-O and then immunostained with VCAM-1 and CD11b antibodies (n = 4). (b–c) Quantification of VCAM-1 and CD11b expressions by IHC score (n = 4). # P < 0.05 compared with controls; *P < 0.05 compared with untreated CIA mice.

Discussion

RA is characterized by immune cell infiltration into the synovium, synovial inflammation, pannus formation, joint trauma, and degradation (37). Activated RASFs in the diseased synovium migrate to non-affected joints via CAMs and favor monocyte recruitment into the areas of inflammation (38, 39). This study revealed higher levels of VCAM-1 and CD11b expressions in human RA synovial tissue compared with healthy control tissue in analyses of GEO database records and IHC staining. A. cinnamomea has long been used by traditional medicine practitioners in Taiwan in the treatment of inflammation, various cancers, and intoxication (4042). Analyses of biological activities of the crude extracts and main bioactive compounds of A. cinnamomea have identified 139 bioactive compounds, including terpenoids, benzenoids, and purine nucleosides, amongst others (43); the main chemical components are ergostane-type triterpenoids, the most abundant of which is Antcin K (44). Our screening of Antcin K and four other ergostane triterpenoids (eburicoic acid, zhankuic acids A and C, and dehydroeburicoic acid) isolated from A. cinnamomea revealed that Antcin K was the most potent anti-inflammatory compound. In our previous investigations, Antcin K exhibited potent anti-inflammatory and anti-angiogenic effects (16, 17) and also prevented joint degradation in CIA mice (16). In this study, Antcin K appeared to inhibit VCAM-1 expression and monocyte adhesion in RASFs. The findings also indicate that the MEK1/2-ERK, p38, and AP-1 signaling pathways mediate the inhibitory effects of Antcin K upon VCAM-1 expression.

VCAM-1 expression is increased during inflammation and is an important mediator of leukocyte-endothelial cell adhesion in inflamed tissue (7), which was supported by the findings of high levels of VCAM-1 expression in RA synovial tissue in this study. It may be that serum VCAM-1 levels in RA are linked to the autoimmune and inflammatory reactions of the disorder and, therefore, reflect the degree of disease severity and predict the consequences of treatment (6). Indeed, inhibiting VCAM-1 expression inhibits the development of CIA and adjuvant-induced arthritis (45). Following Antcin K treatment, our observation of significant reductions in VCAM-1 expression and levels of monocyte adhesion in human RASFs suggest that this compound can effectively inhibit RA disease.

The MEK/ERK and p38 signaling cascades are critical players during inflammatory responses in diseases mediated by metalloproteinases and proinflammatory cytokines (46, 47). Phosphorylation of MEK/ERK signaling is increased by adipokine-induced upregulation of IL-6 expression in human osteoblastic cells (32) and VEGF expression in human RASFs (48). Moreover, the selective MEK1/2 inhibitor, U0126, effectively suppresses proinflammatory cytokine synthesis in LPS-stimulated monocytes (49). Furthermore, high levels of VCAM-1 and proinflammatory cytokine expression in immortalized mouse cardiac endothelial cells (MCECs) depend upon the MEK/ERK and p38 signaling pathways (50). We observed that Antcin K effectively inhibited MEK1/2-ERK and p38 phosphorylation and is, therefore, capable of inhibiting VCAM-1 expression and monocyte adhesion in RASFs.

AP-1 is a key contributor to inflammatory bone diseases (51), and AP-1 proteins regulate inflammatory processes in macrophages by stimulating cytokine secretion (52). Targeting AP-1 activity in RA is worthwhile, (53) as shown, for instance, by findings from a mouse model of RA, in which selective inhibition of AP-1 successfully inhibited disease progression (54). Similarly, the inhibitory effects of soya-cerebroside upon proinflammatory cytokine synthesis in RASFs occur partly through the suppression of AP-1 signaling (26). In this study, Antcin K significantly inhibited c-Jun phosphorylation in human RASFs and attenuated AP-1 luciferase activity.

Monocytes express CD11 integrins that, in turn, express VCAM-1 ligands on the cell surface of the activated endothelium (55). It has been suggested that VCAM-1 plays a crucial role in the retention and survival of infiltrating monocytes in synovial tissue (56). Using TNF blockers to inhibit VCAM-1 expression decreases CAM expression and lowers macrophage counts in synovial tissue (56). We observed that VCAM-1 and CD11b signaling was inhibited in synovial tissue from ankle joints of CIA mice treated with Antcin K (10 mg/kg or 30 mg/kg). A limitation of this study is that the bioavailability of ingested doses of Antcin K remains unknown. We would like to resolve this aspect in future projects.

Conclusions

Our findings revealed that Antcin K dose-dependently inhibits VCAM-1 expression in RASFs and inhibits monocyte adhesion in RASFs via the MEK1/2-ERK, p38, and AP-1 signaling cascades (see Fig. 9). Antcin K may have potential as a therapeutic agent in the management of RA.

Fig 9
Fig. 9. The schema illustrates how the antiarthritic mechanisms of Antcin K are achieved through the inhibition of VCAM-1 via the MEK1/2-ERK, p38, and AP-1 signaling pathways.

Institutional review board statement

The study protocol was approved by the Institutional Review Board (IRB) of China Medical University Hospital (CMUH108-REC3-039).

Informed consent statement

All study participants signed informed written consent forms prior to participation.

Data availability statement

The original data to this present study are available from the corresponding authors.

CRediT authorship contribution statement

Conceptualization, D.A. and C.H.Ta.; methodology, D.A., S.C.L., and Y.Y.L.; software, S.L.C., C.C.H., and C.H.Ts.; validation, D.A., S.C.L., Y.Y.L., and C.Y.K.; formal analysis, D.A. and S.L.C.; investigation, D.A. and C.H.Ta.; resources, C.H.Ta. and Y.H.K.; data curation, D.A., W.C.H., and Y.C.W.; writing – original draft preparation, D.A.; writing – review and editing, C.H.Ta. and D.A.; supervision, C.H.Ta. and S.C.L.; project administration, C.H.Ta.; funding acquisition, C.H.Ta. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

The authors would like to thank Iona J. MacDonald from China Medical University, Taichung, Taiwan, for her editing of this manuscript.

References

  1. Cross M, Smith E, Hoy D, Carmona L, Wolfe F, Vos T, et al. The global burden of rheumatoid arthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis 2014; 73(7): 1316–22. doi: 10.1136/annrheumdis-2013-204627
  2. Shrivastava AK, Pandey A. Inflammation and rheumatoid arthritis. J Physiol Biochem 2013; 69(2): 335–47. doi: 10.1007/s13105-012-0216-5
  3. Smiljanovic B, Radzikowska A, Kuca-Warnawin E, Kurowska W, Grun JR, Stuhlmuller B, et al. Monocyte alterations in rheumatoid arthritis are dominated by preterm release from bone marrow and prominent triggering in the joint. Ann Rheum Dis 2018; 77(2): 300–8. doi: 10.1136/annrheumdis-2017-211649
  4. Fukui S, Iwamoto N, Takatani A, Igawa T, Shimizu T, Umeda M, et al. M1 and M2 monocytes in rheumatoid arthritis: a contribution of imbalance of M1/M2 monocytes to osteoclastogenesis. Front Immunol 2017; 8: 1958. doi: 10.3389/fimmu.2017.01958
  5. Hasseli R, Frommer KW, Schwarz M, Hulser ML, Schreiyack C, Arnold M, et al. Adipokines and inflammation alter the interaction between rheumatoid arthritis synovial fibroblasts and endothelial cells. Front Immunol 2020; 11(925). doi: 10.3389/fimmu.2020.00925
  6. Wang LF, Ding YJ, Guo XQ, Zhao Q. Role and mechanism of vascular cell adhesion molecule-1 in the development of rheumatoid arthritis. Exp Ther Med 2015; 10(3): 1229–33. doi: 10.3892/etm.2015.2635
  7. Kong DH, Kim YK, Kim MR, Jang JH, Lee S. Emerging roles of vascular cell adhesion molecule-1 (VCAM-1) in immunological disorders and cancer. Int J Mol Sci 2018; 19(4): 1057. doi: 10.3390/ijms19041057
  8. Silverman MD, Haas CS, Rad AM, Arbab AS, Koch AE. The role of vascular cell adhesion molecule 1/ very late activation antigen 4 in endothelial progenitor cell recruitment to rheumatoid arthritis synovium. Arthritis Rheum 2007; 56(6): 1817–26. doi: 10.1002/art.22706
  9. Smith MD, Slavotinek J, Au V, Weedon H, Parker A, Coleman M, et al. Successful treatment of rheumatoid arthritis is associated with a reduction in synovial membrane cytokines and cell adhesion molecule expression. Rheumatology 2001; 40(9): 965–77. doi: 10.1093/rheumatology/40.9.965
  10. Oberoi R, Schuett J, Schuett H, Koch AK, Luchtefeld M, Grote K, et al. Targeting tumor necrosis factor-alpha with adalimumab: effects on endothelial activation and monocyte adhesion. PLoS One 2016; 11(7): e0160145. doi: 10.1371/journal.pone.0160145
  11. Benjamin O, Bansal P, Goyal A, Lappin SL. Disease modifying anti-rheumatic drugs (DMARD). StatPearls, Treasure Island (FL): StatPearls Publishing; 2018.
  12. Al-Malaq HM, Al-Arfaj HF, Al-Arfaj AS. Adverse drug reactions caused by methotrexate in Saudi population. Saudi Pharm J 2012; 20(4): 301–5. doi: 10.1016/j.jsps.2012.05.004
  13. Chakraborty D, Gupta K, Biswas S. A mechanistic insight of phytoestrogens used for rheumatoid arthritis: an evidence-based review. Biomed Pharmacother 2021; 133: 111039. doi: 10.1016/j.biopha.2020.111039
  14. Meng JH, Zhang WK, Wang C, Zhang W, Zhou CH, Jiang GY, et al. Catalpol suppresses osteoclastogenesis and attenuates osteoclast- derived bone resorption by modulating PTEN activity. Biochem Pharmacol 2020; 171: 113715. doi: 10.1016/j.bcp.2019.113715
  15. Liu XY, Xu L, Wang Y, Li JX, Zhang Y, Zhang C, et al. Protective effects of total flavonoids of Astragalus against adjuvant-induced arthritis in rats by regulating OPG/RANKL/NF-kappaB pathway. Int Immunopharmacol 2017; 44: 105–14. doi: 10.1016/j.intimp.2017.01.010
  16. Achudhan D, Liu S-C, Lin Y-Y, Huang C-C, Tsai C-H, Ko C-Y, et al. Antcin K inhibits TNF-α, IL-1β and IL-8 expression in synovial fibroblasts and ameliorates cartilage degradation: implications for the treatment of rheumatoid arthritis. Front Immunol 2021; 12(790925). doi: 10.3389/fimmu.2021.790925
  17. Achudhan D, Liu SC, Lin YY, Lee HP, Wang SW, Huang WC, et al. Antcin K inhibits VEGF-dependent angiogenesis in human rheumatoid arthritis synovial fibroblasts. J Food Biochem 2021; 46: e14022. doi: 10.1111/jfbc.14022
  18. Huang CC, Chiou CH, Liu SC, Hu SL, Su CM, Tsai CH, et al. Melatonin attenuates TNF-α and IL-1β expression in synovial fibroblasts and diminishes cartilage degradation: implications for the treatment of rheumatoid arthritis. J Pineal Res 2019; 66(3): e12560. doi: 10.1111/jpi.12560
  19. Lee H-P, Liu S-C, Wang Y-H, Chen B-C, Chen H-T, Li T-M, et al. Cordycerebroside A suppresses VCAM-dependent monocyte adhesion in osteoarthritis synovial fibroblasts by inhibiting MEK/ERK/AP-1 signaling. J Funct Foods 2021; 86: 104712. doi: 10.1016/j.jff.2021.104712
  20. Chen WC, Lin CY, Kuo SJ, Liu SC, Lu YC, Chen YL, et al. Resistin enhances VCAM-1 expression and monocyte adhesion in human osteoarthritis synovial fibroblasts by inhibiting MiR-381 expression through the PKC, p38, and JNK signaling pathways. Cells 2020; 9(6): 1369. doi: 10.3390/cells9061369
  21. Lee KT, Su CH, Liu SC, Chen BC, Chang JW, Tsai CH, et al. Cordycerebroside A inhibits ICAM-1-dependent M1 monocyte adhesion to osteoarthritis synovial fibroblasts. J Food Biochem 2022; 00: e14108. doi: 10.1111/jfbc.14108.
  22. Liu S-C, Tsai C-H, Wu T-Y, Tsai C-H, Tsai F-J, Chung J-G, et al. Soya-cerebroside reduces IL-1β-induced MMP-1 production in chondrocytes and inhibits cartilage degradation: implications for the treatment of osteoarthritis. Food Agric Immunol 2019; 30(1): 620–32. doi: 10.1080/09540105.2019.1611745
  23. Lee HP, Wang SW, Wu YC, Lin LW, Tsai FJ, Yang JS, et al. Soya-cerebroside inhibits VEGF-facilitated angiogenesis in endothelial progenitor cells. Food Agric Immunol 2020; 31(1): 193–204. doi: 10.1080/09540105.2020.1713055
  24. Wang YH, Kuo SJ, Liu SC, Wang SW, Tsai CH, Fong YC, et al. Apelin affects the progression of osteoarthritis by regulating VEGF-dependent angiogenesis and miR-150-5p expression in human synovial fibroblasts. Cells 2020; 9(3): 594. doi: 10.3390/cells9030594
  25. Cheng F-J, Huynh T-K, Yang C-S, Hu D-W, Shen Y-C, Tu C-Y, et al. Hesperidin is a potential inhibitor against SARS-CoV-2 infection. Nutrients 2021; 13(8): 2800. doi: 10.3390/nu13082800
  26. Lee HP, Wu YC, Chen BC, Liu SC, Li TM, Huang WC, et al. Soya-cerebroside reduces interleukin production in human rheumatoid arthritis synovial fibroblasts by inhibiting the ERK, NF-kappa B and AP-1 signalling pathways. Food Agric Immunol 2020; 31(1): 740–50.
  27. Lee H-P, Chen P-C, Wang S-W, Fong Y-C, Tsai C-H, Tsai F-J, et al. Plumbagin suppresses endothelial progenitor cell-related angiogenesis in vitro and in vivo. J Funct Foods 2019; 52: 537–44. doi: 10.1016/j.jff.2018.11.040
  28. Lin YY, Ko CY, Liu SC, Wang YH, Hsu CJ, Tsai CH, et al. miR-144-3p ameliorates the progression of osteoarthritis by targeting IL-1beta: potential therapeutic implications. J Cell Physiol 2021; 236(10): 6988–7000.
  29. Su C-H, Lin C-Y, Tsai C-H, Lee H-P, Lo L-C, Huang W-C, et al. Betulin suppresses TNF-α and IL-1β production in osteoarthritis synovial fibroblasts by inhibiting the MEK/ERK/NF-κB pathway. J Funct Foods 2021; 86: 104729. doi: 10.1016/j.jff.2021.104729
  30. Denys A, Clavel G, Lemeiter D, Schischmanoff O, Boissier MC, Semerano L. Aortic VCAM-1: an early marker of vascular inflammation in collagen-induced arthritis. J Cell Mol Med 2016; 20(5): 855–63. doi: 10.1111/jcmm.12790
  31. Tokuhira M, Hosaka S, Volin MV, Haines III GK, Katschke Jr KJ, Kim S, et al. Soluble vascular cell adhesion molecule 1 mediation of monocyte chemotaxis in rheumatoid arthritis. Arthritis Rheum 2000; 43(5): 1122–33.
  32. Hu S-L, Huang C-C, Tzeng T-T, Liu S-C, Tsai C-H, Fong Y-C, et al. S1P promotes IL-6 expression in osteoblasts through the PI3K, MEK/ERK and NF-κB signaling pathways. Int J Med Sci 2020; 17(9): 1207–14. doi: 10.7150/ijms.44612
  33. Shen Y, Teng L, Qu Y, Liu J, Zhu X, Chen S, et al. Anti-proliferation and anti-inflammation effects of corilagin in rheumatoid arthritis by downregulating NF-κB and MAPK signaling pathways. J Ethnopharmacol 2022; 284: 114791. doi: 10.1016/j.jep.2021.114791
  34. Erusappan T, Paramasivam S, Ekambaram SP. Identification of galangin as the bioactive compound from Alpinia calcarata (Haw.) Roscoe rhizomes to inhibit IRAK-1/ MAPK/ NF-κB p65 and JAK-1 signaling in LPS stimulated RAW 264.7 cells. J Ethnopharmacol 2022; 288: 114975. doi: 10.1016/j.jep.2022.114975
  35. Le Rossignol S, Ketheesan N, Haleagrahara N. Redox-sensitive transcription factors play a significant role in the development of rheumatoid arthritis. Int Rev Immunol 2018; 37(3): 129–43. doi: 10.1080/08830185.2017.1363198
  36. Sawa Y, Ueki T, Hata M, Iwasawa K, Tsuruga E, Kojima H, et al. LPS-induced IL-6, IL-8, VCAM-1, and ICAM-1 expression in human lymphatic endothelium. J Histochem Cytochem 2008; 56(2): 97–109.
  37. Guo Q, Wang Y, Xu D, Nossent J, Pavlos NJ, Xu J. Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies. Bone Res 2018; 6: 15. doi: 10.1038/s41413-018-0016-9
  38. Neumann E, Lefevre S, Zimmermann B, Geyer M, Lehr A, Umscheid T, et al. Migratory potential of rheumatoid arthritis synovial fibroblasts: additional perspectives. Cell Cycle 2010; 9(12): 2286–91.
  39. Lefevre S, Knedla A, Tennie C, Kampmann A, Wunrau C, Dinser R, et al. Synovial fibroblasts spread rheumatoid arthritis to unaffected joints. Nat Med 2009; 15(12): 1414–20. doi: 10.1038/nm.2050
  40. Kumar K, Wang S-Y. Antioxidant properties of Antrodia cinnamomea: an extremely rare and coveted medicinal mushroom endemic to Taiwan. In: Medicinal plants and fungi: recent advances in research and development. Singapore: Springer 2017, pp. 135–64.
  41. Huang TT, Wu SP, Chong KY, Ojcius DM, Ko YF, Wu YH, et al. The medicinal fungus Antrodia cinnamomea suppresses inflammation by inhibiting the NLRP3 inflammasome. J Ethnopharmacol 2014; 155(1): 154–64. doi: 10.1016/j.jep.2014.04.053
  42. Ganesan N, Baskaran R, Velmurugan BK, Thanh NC. Antrodia cinnamomea-An updated minireview of its bioactive components and biological activity. J Food Biochem 2019; 43(8): e12936. doi: 10.1111/jfbc.12936
  43. Zhang Y, Lv P, Ma J, Chen N, Guo H, Chen Y, et al. Antrodia cinnamomea exerts an anti-hepatoma effect by targeting PI3K/AKT-mediated cell cycle progression in vitro and in vivo. Acta Pharm Sin B 2022; 12(2): 890–906. doi: 10.1016/j.apsb.2021.07.010
  44. Huang YL, Chu YL, Ho CT, Chung JG, Lai CI, Su YC, et al. Antcin K, an active triterpenoid from the fruiting bodies of basswood-cultivated antrodia cinnamomea, inhibits metastasis via suppression of lntegrin-mediated adhesion, migration, and invasion in human hepatoma cells. J Agr Food Chem 2015; 63(18): 4561–9. doi: 10.1021/jf5059304
  45. Carter RA, Campbell IK, O’Donnel KL, Wicks IP. Vascular cell adhesion molecule-1 (VCAM-1) blockade in collagen-induced arthritis reduces joint involvement and alters B cell trafficking. Clin Exp Immunol 2002; 128(1): 44–51. doi: 10.1046/j.1365-2249.2002.01794.x
  46. Thiel MJ, Schaefer CJ, Lesch ME, Mobley JL, Dudley DT, Tecle H, et al. Central role of the MEK/ERK MAP kinase pathway in a mouse model of rheumatoid arthritis: potential proinflammatory mechanisms. Arthritis Rheum 2007; 56(10): 3347–57. doi: 10.1002/art.22869
  47. Yang Y, Kim SC, Yu T, Yi YS, Rhee MH, Sung GH, et al. Functional roles of p38 mitogen-activated protein kinase in macrophage-mediated inflammatory responses. Mediators Inflamm 2014; 2014: 352371. doi: 10.1155/2014/352371
  48. Huang CC, Law YY, Liu SC, Hu SL, Lin JA, Chen CJ, et al. Adiponectin promotes VEGF expression in rheumatoid arthritis synovial fibroblasts and induces endothelial progenitor cell angiogenesis by inhibiting miR-106a-5p. Cells 2021; 10(10): 2627. doi: 10.3390/cells10102627
  49. Li L, Li Y, Shi D, Liu H, Wang B, Sun Y. Xuebijing inhibit inflammation, oxidative stress and promote apoptosis in human synovial cells via inhibition of MEK1/2 and NF-κB pathway. Isr J Plant Sci 2021; 1(aop): 1–9. doi: 10.1163/22238980-bja10032
  50. Singh RJ, Mason JC, Lidington EA, Edwards DR, Nuttall RK, Khokha R, et al. Cytokine stimulated vascular cell adhesion molecule-1 (VCAM-1) ectodomain release is regulated by TIMP-3. Cardiovasc Res 2005; 67(1): 39–49. doi: 10.1016/j.cardiores.2005.02.020
  51. Wagner EF, Eferl R. Fos/AP-1 proteins in bone and the immune system. Immunol Rev 2005; 208: 126–40. doi: 10.1111/j.0105-2896.2005.00332.x
  52. Hannemann N, Jordan J, Paul S, Reid S, Baenkler HW, Sonnewald S, et al. The AP-1 transcription factor c-Jun promotes arthritis by regulating cyclooxygenase-2 and arginase-1 expression in macrophages. J Immunol 2017; 198(9): 3605–14. doi: 10.4049/jimmunol.1601330
  53. Zenz R, Eferl R, Scheinecker C, Redlich K, Smolen J, Schonthaler HB, et al. Activator protein 1 (Fos/Jun) functions in inflammatory bone and skin disease. Arthritis Res Ther 2008; 10(1): 201. doi: 10.1186/ar2338
  54. Aikawa Y, Morimoto K, Yamamoto T, Chaki H, Hashiramoto A, Narita H, et al. Treatment of arthritis with a selective inhibitor of c-Fos/activator protein-1. Nat Biotechnol 2008; 26(7): 817–23. doi: 10.1038/nbt1412
  55. Ruiz-Limon P, Ortega-Castro R, Barbarroja N, Perez-Sanchez C, Jamin C, Patino-Trives AM, et al. Molecular characterization of monocyte subsets reveals specific and distinctive molecular signatures associated with cardiovascular disease in rheumatoid arthritis. Front Immunol 2019; 10: 1111. doi: 10.3389/fimmu.2019.01111
  56. Herenius MM, Thurlings RM, Wijbrandts CA, Bennink RJ, Dohmen SE, Voermans C, et al. Monocyte migration to the synovium in rheumatoid arthritis patients treated with adalimumab. Ann Rheum Dis 2011; 70(6): 1160–2. doi: 10.1136/ard.2010.141549