Gambogic acid suppresses inflammation in rheumatoid arthritis rats via PI3K/Akt/mTOR signaling pathway

  • Authors:
    • Xiaodan Wu
    • Li Long
    • Jian Liu
    • Jin Zhang
    • Tong Wu
    • Xixi Chen
    • Bing Zhou
    • Ting‑Zhuo Lv
  • View Affiliations

  • Published online on: September 11, 2017     https://doi.org/10.3892/mmr.2017.7459
  • Pages: 7112-7118
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gamboge is the dried resin secreted by the Garcinia maingayi gambogic tree and is a substance that may be used to treat a variety of diseases, exhibits anti‑tumor and detoxification effects and prevents bleeding. The primary active constituent is gambogic acid. The present study aimed to investigate the anti‑inflammatory effects of gambogic acid in rheumatoid arthritis (RA) rats and to elucidate the mechanisms by which these effects occur. The swelling degree, the clinical arthritic scoring and pain threshold measurements were used to evaluate the effects of gambogic acid on RA. ELISA kits and western blot analysis were used to investigate inflammatory processes and the expression of RA‑associated proteins, respectively. The present results demonstrated that gambogic acid significantly inhibited the degree of right foot swelling, increased pain thresholds and reduced clinical arthritic scores of RA rats. Treatment with gambogic acid suppressed the activities of interleukin (IL)‑1β and IL‑6, promoted the protein expression of phosphorylated (p)‑Akt serine/threonine kinase (Akt), p‑mammalian target protein of rapamycin (mTOR) and inhibited hypoxia‑inducible factor‑1α and vascular endothelial growth factor expression in RA rats. The results of the present study therefore suggest that the anti‑inflammatory effects of gambogic acid in RA rats occur via regulation of the phosphoinositide 3‑kinase/Akt/mTOR signaling pathway.

Introduction

Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease with etiology that remains to be fully elucidated. At present, it is widely recognized that RA is an autoimmune disease. The disease is often presented in facet joints, including hands, wrists and feet with symmetric distribution (1). Patients at an early stage of the disease, often present with pain in red, swelled and heated joints and functional barriers, whereas patients at an advanced stage of the disease present ankylosis or malformation at varying degrees, accompanied with atrophy of bone muscle, bone necrosis and numerous disabling symptoms (2). The specific pathogenesis of the disease is not clear and may be associated with multiple factors, including social differences, geographical conditions, internal secretion alterations, vocational status, nutrition and metabolic disturbance and bacterial and viral infections (3). RA as of yet has no effective treatment and its therapeutic regimen primarily involves relieving pain and inflammation (4).

Arthrocele at early stage results from synovium congestion, edema and exudation of articular cavity, in addition to periarticular tissue edema (5). In rheumatic synovitis, fenestra expansion of synovial endothelial cells and stimulation of inflammatory mediators, including histamine, bradykinin, prostaglandin E2, free radicals and neurosecretion of substance P result in hemangiectasis, increase in capillary permeability and exudation, which lead to synovial membrane and periarticular tissue edema (6). Collagen and protein polysaccharide degradation are mediated by cytokines interleukin (IL)-1, IL-6, IL-8 and tumor necrosis factor (TNF)-α, chondrocyte swelling and degeneration are mediated by nitric oxide (NO) and NO-induced oxygenase expression in cartilage (5,7). Adhesion of hemameba, T cells, cell endothelial cells and fibrinogen mediated by adhesion molecules promote formation of a fibrin clot, resulting in disturbed blood vessel function and a subsequent synovium inflammatory response (8). Growth hormone inhibiting factor, TNF-α and IL increase in synovium results in increasing T cell expression with autoantigen reactivity, resulting in arthritic symptoms (9).

The phosphoinositide 3-kinase (PI3K), Akt serine/threonine kinase (Akt) signaling pathway is an important intracellular transduction pathway and has previously been demonstrated to be associated with the development of RA (10). The active state of the signaling pathway is tightly regulated by the negative regulatory factor phosphatase and tensin homolog (PTEN), and the regulatory factors TNF-α, transforming growth factor (TGF)-β and TNF-related apoptosis-inducing ligand (11). In the synovial cells of RA patients, the signaling pathway is maintained in the abnormal activated state, resulting in high expression of downstream anti-apoptosis genes and a subsequent impact on multiple downstream effector molecules (12). Furthermore, it is important in the synovial cell proliferation and apoptotic imbalance of RA patient joints. Excessively-grown synovial cells infiltrate and grow in the cartilage articularis and bone tissues, resulting in joint deformity and dysfunction. Therefore, the inhibition of the PI3K-AKT signal pathway may negatively regulate proteins, reverse excessive proliferation of RA synoviocytes, and may provide a novel target for curing the disease (13).

The primary constituents of gamboge are composed of 70–80% resin, 15–25% gum and include gambogic acid, neogambogic acid, allogambogic acid, morellin, isomorellin, morellic acid and isomorellic constituents. Gambogic acid is the primary effective constituent (14). The pharmacological and pharmacokinetic properties of preparation of gambogic acid have previously been studied (15). The anti-tumor effects of gambogic acid involve induction of tumor cell apoptosis, restraining the cell cycle and impacting oncogenes, cancer suppressor genes and expression of associated proteins (16). The present study therefore aimed to investigate the anti-inflammatory effects of gambogic acid in RA rats and to reveal the potential signaling pathways involved.

Materials and methods

Experimental rat model

All study protocols employed were in accordance with the guidelines of the Animal Care and Use Committee of Baodi District People's Hospital of Tianjin City (Tianjin, China). The study was approved by the Ethics Committee of Sichuan Provincial People Hospital (Chengtu, China). A total of 30 male Sprague-Dawley rats (age, 8–10 weeks; weight, 250–300 g) were obtained from Laboratory Animal Center of Tianjin Medical University (Tianjin, China) and maintained in individual cages under standard conditions (temperature, 22–23°C; humidity, 55–60%; 12-h light/dark cycle), and provided with free access to food and water.

Grouping and model establishment

The experimental rats were randomly divided into 5 groups (6 rats/group): Sham, RA model, gambogic acid (1 mg/kg/day), gambogic acid (5 mg/kg/day) and gambogic acid (10 mg/kg/day) groups for 3 weeks. The chemical structure of gambogic acid (≥95%, high performance liquid chromatography) is indicated in Fig. 1 and was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). In the RA model, gambogic acid (1 mg/kg/day), gambogic acid (5 mg/kg/day) and gambogic acid (10 mg/kg/day) groups, rats were anesthetized with an intraperitoneal injection of 40 mg/kg sodium pentobarbital (Sigma-Aldrich; Merck KGaA). A total of 10 mg/ml Freund's complete adjuvant (Sigma-Aldrich; Merck KGaA) was subcutaneously injected into the 2nd and 3rd toes of the right foot. The right ankle then demonstrated acute inflammatory swelling.

Swelling degree measurement

The posterior right limb was inserted in 20 ml water, and the volume of displaced water was measured as representative of foot volume. The swelling degree was measured as follows: (Foot volume 7 days following RA induction-foot volume prior to RA induction)/(foot volume prior to RA induction)x100%.

Clinical arthritic scoring measurement

The rats were observed according to a macroscopic scoring system, as follows: 0, no sign of arthritis; 1–5, 2 joints involved; 6–10, >2 joints involved; 11–15; severe arthritis of the entire paw and digits. Clinical arthritic scoring was performed three times in each session.

Pain threshold measurement

Pain threshold measurement was conducted as previously described (17). The pressure pain threshold (g) was detected using an electronic pressure pain detector (Somedic AB, Sösdala, Sweden). Pain threshold measurement was conducted three times in each session.

Inflammatory effects measurement

Following sacrifice of rats using 100 mg/kg sodium pentobarbital (Sigma-Aldrich; Merck KGaA), blood was extracted from the inferior vena cava (3 ml) and centrifuged at 3,000 × g for 10 min at 4°C. The supernatant was used to analyze IL-1β (cat. no. ER008-96) and IL-6 (cat. no. ER003-96) activities with ELISA kits, according to the manufacturer's protocol (Genetimes Technology, Inc., Shanghai, China).

Western blotting

The synovium was digested with radioimmunoprecipitation assay lysis buffer (Roche Diagnostics, Basel, Switzerland) at 4°C for 30 min and centrifuged at 12,000 × g for 10 min at 4°C. The supernatant was used to analyze the protein content using a bicinchoninic acid kit (Fermentas; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Equal amounts of extracted protein samples (50 µg) were separated by 10% SDS-PAGE and transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Membranes were blocked using 5% skim milk powder in TBS containing 0.1% Tween-20 at 37°C for 1 h and incubated with the following primary antibodies at 4°C overnight: Anti-Akt (1:300; cat. no. sc-8312), anti-phosphorylated (p)-Akt (1:500; cat. no. sc-7985-R), anti-mammalian target protein of rapamycin (mTOR; 1:300; cat. no. sc-8319), anti-p-mTOR (1:500; cat. no. sc-101738), anti-vascular endothelial growth factor (VEGF; 1:300; cat. no. sc-13083), anti-hypoxia-inducible factor-1α (HIF-1α; 1:300; cat. no. sc-10790) and anti-GAPDH (1:300; cat. no. sc-367714), all obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Membranes were then incubated with goat anti-rabbit immunoglobulin G (1:2,000; cat. no. 14708; Cell Signaling Technology, Inc., Danvers, MA, USA) at 37°C for 1 h. Protein bands were visualized using enhanced chemiluminescence (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Blots were semi-quantified using ImageJ software version 1.41 (National Institutes of Health, Bethesda, MD, USA).

Statistical analysis

Data are expressed as the mean ± standard deviation of 3 independent experiments. One-way analysis of variance followed by the Tukey's post hoc test was used to determine the differences among groups using SPSS software, version 11.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Gambogic acid decreases degree of right foot swelling

The constitutional formula of gambogic acid is presented in Fig. 1. The present study firstly examined the potential effects of gambogic acid on the degree of right foot swelling in RA rats. The degree of right foot swelling was increased in the RA model group compared with sham group (Fig. 2). The results demonstrated that treatment with gambogic acid at 10 mg/kg/day significantly inhibited the degree of right foot swelling in RA rats (Fig. 2).

Gambogic acid decreases clinical arthritic score of RA rats

The potential effects of gambogic acid on the clinical arthritic score of RA rats was next investigated. As presented in Fig. 3, the clinical arthritic score of the RA rat model group significantly increased over the period of 3 weeks, compared with sham group. However, administration of 10 mg/kg/day gambogic acid significantly reduced the clinical arthritic score of RA rats, over this time period (Fig. 3).

Gambogic acid increases pain threshold of the RA rat model

To elucidate the potential effects of gambogic acid on the RA rat model, the pain threshold of the RA rat model was measured. As presented in Fig. 4, compared with sham group, the pain threshold of the RA rat model was decreased. Treatment with gambogic acid significantly increased the pain threshold of the RA rats (Fig. 4).

Gambogic acid decreases inflammation of the RA rat model

To elucidate the anti-inflammatory effect of gambogic acid in the RA rat model, IL-1β and IL-6 activities were analyzed using ELISA kits. Notably, RA significantly promoted IL-1β and IL-6 activities in the RA rat model, compared with sham group (Fig. 5). Following treatment with gambogic acid, the promotion of IL-1β and IL-6 activities in RA rats was significantly suppressed (Fig. 5).

Gambogic acid suppresses inhibition of the Akt signaling pathway in the RA rat model

p-Akt expression was examined from tissue via western blotting, in order to analyze the potential effects of gambogic acid on RA. The p-Akt protein expression was significantly inhibited in the RA rat model, compared with the sham group (Fig. 6). Notably, gambogic acid significantly suppressed the inhibition of p-Akt protein expression in RA rats (Fig. 6).

Gambogic acid activates mTOR signaling pathway in the RA rat model

To explore if gambogic acid activates the mTOR signaling pathway in RA rats, the models were treated with differing doses. The p-mTOR protein expression levels in the RA rat model group were decreased compared with sham group (Fig. 7). However, treatment with gambogic acid significantly activated the mTOR signaling pathway via increased p-mTOR protein expression in RA rats (Fig. 7).

Gambogic acid inhibits VEGF signaling pathway in the RA rat model

To examine the role of the VEGF signaling pathway in the potential effects of gambogic acid on RA, VEGF protein expression was measured using western blotting. As presented in Fig. 8, the activation of VEGF protein expression was significantly increased in the RA model group, compared with sham group. Gambogic acid significantly suppressed the activation of VEGF protein expression in RA rats (Fig. 8).

Gambogic acid inhibits HIF-1α signaling pathway in the RA rat model

To investigate the role of the HIF-1α signaling pathway in mediating the potential effects of gambogic acid on RA, HIF-1α protein expression was measured via western blotting. There was a significant increase in HIF-1α protein expression in the RA rat model group, compared with sham group (Fig. 9). Correspondingly, treatment with gambogic acid significantly suppressed the activation of HIF-1α protein expression in RA rats (Fig. 9).

Discussion

RA is a systemic autoimmune disease with a primary manifestation of corrosive arthritis. The predominant pathological alterations are chronic inflammation of synovial tissues (18). Lesions violate multiple joints of body, generally beginning with facet joints, including hands and feet, present symmetry, and may additionally result in systematic diseases, including rheumatoid, vasculitis and pericarditis (19). Mortality rates are positively correlated with age. The disability rate of the disease is high, having a strong impact on physical and psychological health and daily quality of life (20). At present, anti-inflammatory drugs, slow acting anti-rheumatic drugs, glucocorticoids and biological agents are used as therapeutic agents in clinics, however long-term application of these drugs may result in serious gastrointestinal symptoms and damage of the liver and kidney (21,22). In the present study, gambogic acid significantly inhibited the degree of right foot swelling, reduced the clinical arthritic score of RA and increased the pain threshold of the RA rats. These results suggest that gambogic acid may act as a potential future therapeutic reagent in the treatment of RA.

At the later stages of RA, cartilago articularis and bone tissues of patients are seriously corroded and ultimately result in damage and functional loss of the entire joint structure (9). RA affects ~1% of the total population worldwide (23). It is reported that inflammatory cells and chemotactic factors are important in the development and progression of RA and autoimmune diseases. TNF-α, IL-1β and IL-6 are important in mediating RA-associated mortality, the synovial inflammatory response and the process of osteoclasia (21). At present, inhibitors of IL-1β and IL-6 have already been used for clinical research and therapy of RA and exhibit beneficial effects (24). The findings of the present study revealed that gambogic acid significantly suppressed the promotion of IL-1β and IL-6 activities in RA rats.

PI3K/Akt signaling regulates proliferation and differentiation of B lymphocytes by activating mTOR (12). mTOR is the target molecule of sirolimus and is vital in the regulation of cell growth and proliferation. mTOR forms two types of complexes (C) with different functions, including mTORCl and mTORC2 (11). mTORC1 is sensitive to sirolimus, whereas mTORC2 is not sensitive to sirolimus. mTORC1 reinforces protein synthesis via phosphorylation of 4E-binding protein 1 and ribosomal protein S6 kinase B1. mTOR signaling impacts the cell cycle, cell growth and cell proliferation (25). In numerous human cancers, various important tumor-inhibiting factors (PTEN, tuberous sclerosis 1/2, liver kinase B1) in the mTOR signaling pathway are deficient. Cell mutation and gene amplification in PI3CA (P110α subtype of PI3K), in addition to an Akt activated mutation, result in increases in cell proliferation (13). The present study demonstrated that gambogic acid significantly prevented the decrease in p-Akt and p-mTOR protein expression induced by RA in the rat models. Activation of the PI3K/Akt/mTOR signaling pathway was important in the effect of gambogic acid on RA. Liu et al (14) suggested that gambogic acid induces G0/G1 cell cycle arrest via the PI3K/Akt/mTOR signaling pathway.

Vascular endothelial growth factor is additionally termed vascular permeability enhancement factor (26). It participates in human embryonic development, wound healing, hair growth and diabetic retinopathy, with pathological and physiological roles. The molecular weight of VEGF is ~46 kDa. It is a dimer glycoprotein molecule comprised of two monomers combined, and has two important physiological functions (27). Pro-blood vessel endothelium proliferation is the leading function of VEGF. It is an endothelial cell mitogen and promotes fission of endothelial cells in order to promote neovascularization. The VEGF receptor is located in vascular endothelial cells (28). Therefore, the function of VEGF is concentrated and has stronger specificity (27). VEGF may additionally increase permeability of blood capillaries and is the blood vessel anti-reflection molecule that exhibits the strongest biological effect, >5 million times more potent compared with histamine substances (28). The results of the present study suggested that gambogic acid significantly suppressed the activation of VEGF protein expression, blocking activation of the VEGF signaling pathway in RA rats. Lu et al (16) suggested that gambogic acid may be a structurally novel angiogenesis inhibitor via suppressing VEGF production.

HIF-1α is an important regulatory factor of histiocyte anoxia. RA intra-articular anoxia results in an upregulation of HIF-1α expression in synovial tissues and regulates multiple signaling pathways, including inflammation, angiogenesis, cell invasion and proliferation, and intensifies the occurrence and development of RA (29). In the pathological state, HIF-1α expression is abnormal. The RA intra-articular environment is an anaerobic environment which leads to high expression of HIF-1α in synovial tissues (30). High expression of HIF-1α may promote RA in synovial tissues, secrete chemotactic factors, recruit monocytes, T and B lymphocytes, reinforce differentiation of T helper 17 cells directly and participate in the overall inflammatory response of RA (31). Furthermore, HIF-1α may additionally promote VEGF expression and the subsequent generation of blood vessels (31). In the present study, gambogic acid significantly suppressed the activation of HIF-1α protein expression in RA rats. Lu et al (16) demonstrated that gambogic acid inhibited angiogenesis via suppression of the HIF-1α pathway. These findings suggested that gambogic acid downregulates HIF-1α, in treatment of RA.

In conclusion, the present study demonstrated that gambogic acid significantly inhibited the degree of right foot swelling, increased pain threshold, reduced clinical arthritic score and suppressed inflammation in RA rats, via regulation of the Akt, mTOR, VEGF and HIF-1α signaling pathways. Therefore, gambogic acid may act as a potential future therapeutic reagent in the treatment for RA.

References

1 

Meyfroidt S, Stevens J, De Lepeleire J, Westhovens R, De Cock D, Van Der Elst K, Vanhaecht K and Verschueren P: A general practice perspective on early rheumatoid arthritis management: A qualitative study from Flanders. Eur J Gen Pract. 21:231–237. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Abram SG, Nicol F, Hullin MG and Spencer SJ: The long-term outcome of uncemented low contact stress total knee replacement in patients with rheumatoid arthritis: Results at a mean of 22 years. Bone Joint J. 95-B:1–1499. 2013. View Article : Google Scholar

3 

Avina-Zubieta JA, Abrahamowicz M, De Vera MA, Choi HK, Sayre EC, Rahman MM, Sylvestre MP, Wynant W, Esdaile JM and Lacaille D: Immediate and past cumulative effects of oral glucocorticoids on the risk of acute myocardial infarction in rheumatoid arthritis: A population-based study. Rheumatology (Oxford). 52:68–75. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Halabi H, Alarfaj A, Alawneh K, Alballa S, Alsaeid K, Badsha H, Benitha R, Bouajina E, Al Emadi S, El Garf A, et al: Challenges and opportunities in the early diagnosis and optimal management of rheumatoid arthritis in Africa and the Middle East. Int J Rheum Dis. 18:268–275. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Andersson U and Tracey KJ: HMGB1 as a mediator of necrosis-induced inflammation and a therapeutic target in arthritis. Rheum Dis Clin North Am. 30:627–637, xi. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Szabo-Taylor KE, Eggleton P, Turner CA, Faro ML, Tarr JM, Tóth S, Whiteman M, Haigh RC, Littlechild JA and Winyard PG: Lymphocytes from rheumatoid arthritis patients have elevated levels of intracellular peroxiredoxin 2 and a greater frequency of cells with exofacial peroxiredoxin 2, compared with healthy human lymphocytes. Int J Biochem Cell Biol. 44:1223–1231. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Calmon-Hamaty F, Combe B, Hahne M and Morel J: Lymphotoxin alpha stimulates proliferation and pro-inflammatory cytokine secretion of rheumatoid arthritis synovial fibroblasts. Cytokine. 53:207–214. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Hsiao HB, Hsieh CC, Wu JB, Lin H and Lin WC: Kinsenoside inhibits the inflammatory mediator release in a type-II collagen induced arthritis mouse model by regulating the T cells responses. BMC Complement Altern Med. 16:802016. View Article : Google Scholar : PubMed/NCBI

9 

Pavelka K, Kavanaugh AF, Rubbert-Roth A and Ferraccioli G: Optimizing outcomes in rheumatoid arthritis patients with inadequate responses to disease-modifying anti-rheumatic drugs. Rheumatology (Oxford). 51 Suppl 5:v12–v21. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Yuan H, Yang P, Zhou D, Gao W, Qiu Z, Fang F, Ding S and Xiao W: Knockdown of sphingosine kinase 1 inhibits the migration and invasion of human rheumatoid arthritis fibroblast-like synoviocytes by down-regulating the PI3K/AKT activation and MMP-2/9 production in vitro. Mol Biol Rep. 41:5157–5165. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Mitra A, Raychaudhuri SK and Raychaudhuri SP: IL-22 induced cell proliferation is regulated by PI3K/Akt/mTOR signaling cascade. Cytokine. 60:38–42. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Chiu YC, Lin CY, Chen CP, Huang KC, Tong KM, Tzeng CY, Lee TS, Hsu HC and Tang CH: Peptidoglycan enhances IL-6 production in human synovial fibroblasts via TLR2 receptor, focal adhesion kinase, Akt, and AP-1-dependent pathway. J Immunol. 183:2785–2792. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Li GQ, Zhang Y, Liu D, Qian YY, Zhang H, Guo SY, Sunagawa M, Hisamitsu T and Liu YQ: PI3 Kinase/Akt/HIF-1α pathway is associated with hypoxia-induced epithelial-mesenchymal transition in fibroblast-like synoviocytes of rheumatoid arthritis. Mol Cell Biochem. 372:221–231. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Liu Y, Li W, Ye C, Lin Y, Cheang TY, Wang M, Zhang H and Wang S, Zhang L and Wang S: Gambogic acid induces G0/G1 cell cycle arrest and cell migration inhibition via suppressing PDGF receptor β tyrosine phosphorylation and Rac1 activity in rat aortic smooth muscle cells. J Atheroscler Thromb. 17:901–913. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Jang SW, Okada M, Sayeed I, Xiao G, Stein D, Jin P and Ye K: Gambogic amide, a selective agonist for TrkA receptor that possesses robust neurotrophic activity, prevents neuronal cell death. Proc Natl Acad Sci USA. 104:16329–16334. 2007; View Article : Google Scholar : PubMed/NCBI

16 

Lu N, Yang Y, You QD, Ling Y, Gao Y, Gu HY, Zhao L, Wang XT and Guo QL: Gambogic acid inhibits angiogenesis through suppressing vascular endothelial growth factor-induced tyrosine phosphorylation of KDR/Flk-1. Cancer Lett. 258:80–89. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Jia Z and He J: Paeoniflorin ameliorates rheumatoid arthritis in rat models through oxidative stress, inflammation and cyclooxygenase 2. Exp Ther Med. 11:655–659. 2016. View Article : Google Scholar : PubMed/NCBI

18 

de la Torre I, Leandro MJ, Edwards JC and Cambridge G: Baseline serum immunoglobulin levels in patients with rheumatoid arthritis: Relationships with clinical parameters and with B-cell dynamics following rituximab. Clin Exp Rheumatol. 30:554–560. 2012.PubMed/NCBI

19 

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

20 

El-Kady IM and El-Masry SA: Pro-inflammatory and anti-inflammatory cytokines profile in rheumatoid arthritis patients. Egypt J Immunol. 15:109–114. 2008.PubMed/NCBI

21 

Ferraccioli G and Gremese E: Thrombogenicity of TNF alpha in rheumatoid arthritis defined through biological probes: TNF alpha blockers. Autoimmun Rev. 3:261–266. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Liu D, Guo M, Hu Y, Liu T, Yan J, Luo Y, Yun M, Yang M, Zhang J and Guo L: Effect of sanhuangwuji powder, anti-rheumatic drugs, and ginger-partitioned acupoint stimulation on the treatment of rheumatoid arthritis with peptic ulcer: A randomized controlled study. J Tradit Chin Med. 35:273–280. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Cobo-Ibáñez T, Descalzo MÁ, Loza-Santamaría E, Carmona L and Munoz-Fernandez S: Serious infections in patients with rheumatoid arthritis and other immune-mediated connective tissue diseases exposed to anti-TNF or rituximab: Data from the Spanish registry BIOBADASER 2.0. Rheumatol Int. 34:953–961. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Xu Y, Zhu Q, Song J, Liu H, Miao Y, Yang F, Wang F, Cheng W, Xi Y, Niu X, et al: Regulatory effect of iguratimod on the balance of Th subsets and inhibition of inflammatory cytokines in patients with rheumatoid arthritis. Mediators Inflamm. 2015:3560402015. View Article : Google Scholar : PubMed/NCBI

25 

Wu T and Mohan C: The AKT axis as a therapeutic target in autoimmune diseases. Endocr Metab Immune Disord Drug Targets. 9:145–150. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Oranskiy SP, Yeliseyeva LN, Tsanaeva AV and Zaytseva NV: Body composition and serum levels of adiponectin, vascular endothelial growth factor, and interleukin-6 in patients with rheumatoid arthritis. Croat Med J. 53:350–356. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Hetland ML, Christensen IJ, Lottenburger T, Johansen JS, Svendsen MN, Hørslev-Petersen K, Nielsen L and Nielsen HJ: Circulating VEGF as a biological marker in patients with rheumatoid arthritis? Preanalytical and biological variability in healthy persons and in patients. Dis Markers. 24:1–10. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Hah YS, Koh YJ, Lim HS, Kim HO, Cheon YH, Noh HS, Jang KY, Lee SY, Lee GM, Koh GY and Lee SI: Double-antiangiogenic protein DAAP targeting vascular endothelial growth factor A and angiopoietins attenuates collagen-induced arthritis. Arthritis Res Ther. 15:R852013. View Article : Google Scholar : PubMed/NCBI

29 

Li GF, Qin YH and Du PQ: Andrographolide inhibits the migration, invasion and matrix metalloproteinase expression of rheumatoid arthritis fibroblast-like synoviocytes via inhibition of HIF-1α signaling. Life Sci. 136:67–72. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Brouwer E, Gouw AS, Posthumus MD, van Leeuwen MA, Boerboom AL, Bijzet J, Bos R, Limburg PC, Kallenberg CG and Westra J: Hypoxia inducible factor-1-alpha (HIF-1alpha) is related to both angiogenesis and inflammation in rheumatoid arthritis. Clin Exp Rheumatol. 27:945–951. 2009.PubMed/NCBI

31 

Seo Y, Ji YW, Lee SM, Shim J, Noh H, Yeo A, Park C, Park MS, Chang EJ and Lee HK: Activation of HIF-1α (hypoxia inducible factor-1α) prevents dry eye-induced acinar cell death in the lacrimal gland. Cell Death Dis. 5:e13092014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 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
Wu X, Long L, Liu J, Zhang J, Wu T, Chen X, Zhou B and Lv TZ: Gambogic acid suppresses inflammation in rheumatoid arthritis rats via PI3K/Akt/mTOR signaling pathway. Mol Med Rep 16: 7112-7118, 2017
APA
Wu, X., Long, L., Liu, J., Zhang, J., Wu, T., Chen, X. ... Lv, T. (2017). Gambogic acid suppresses inflammation in rheumatoid arthritis rats via PI3K/Akt/mTOR signaling pathway. Molecular Medicine Reports, 16, 7112-7118. https://doi.org/10.3892/mmr.2017.7459
MLA
Wu, X., Long, L., Liu, J., Zhang, J., Wu, T., Chen, X., Zhou, B., Lv, T."Gambogic acid suppresses inflammation in rheumatoid arthritis rats via PI3K/Akt/mTOR signaling pathway". Molecular Medicine Reports 16.5 (2017): 7112-7118.
Chicago
Wu, X., Long, L., Liu, J., Zhang, J., Wu, T., Chen, X., Zhou, B., Lv, T."Gambogic acid suppresses inflammation in rheumatoid arthritis rats via PI3K/Akt/mTOR signaling pathway". Molecular Medicine Reports 16, no. 5 (2017): 7112-7118. https://doi.org/10.3892/mmr.2017.7459