Elsevier

International Immunopharmacology

Volume 52, November 2017, Pages 34-43
International Immunopharmacology

Alpha-Mangostin protects rat articular chondrocytes against IL-1β-induced inflammation and slows the progression of osteoarthritis in a rat model

https://doi.org/10.1016/j.intimp.2017.08.010Get rights and content

Highlights

  • α-MG inhibited IL-1β-induced inflammatory cytokine production in rat chondrocytes.

  • α-MG could suppress the activation NF-κB signaling.

  • α-MG prevented cartilage degradation in OA rat model.

  • α-MG may have a potential therapeutic value for OA.

Abstract

Osteoarthritis (OA) is a joint disease characterized by inflammation and cartilage degradation. α-Mangostin (α-MG), which can be isolated from the fruit of the tropical evergreen tree Garcinia mangostana-L, is known to have anti-inflammatory properties. The aim of the study was to investigate the use of α-MG in the treatment of OA, using both rat chondrocytes and an OA rat model induced by destabilization of the medial meniscus (DMM). Rat chondrocytes were pretreated with α-MG (0, 1.25, 2.5, and 5.0 μg/ml for 24 h) prior to stimulation with interleukin-1β (IL-1β) (10 ng/ml for 24 h). Nitric oxide (NO) production was determined using the Griess method and prostaglandin E2 (PGE2) was assessed using an enzyme-linked immunosorbent assay (ELISA). The expression of inducible nitric oxide synthase (INOS), cyclooxygenase-2 (COX-2), matrix metalloproteinase-3, -9, and -13 (MMP-3, MMP-9, and MMP-13), Collagen II, and Aggrecan were detected by both quantitative real-time PCR (qRT-PCR) and a western blot analysis. Nuclear factor-κB (NF-κB) signaling molecules were detected by western blot analysis. Detection of p65 nuclear translocation of NF-κB was examined using immunofluorescence staining. The OA rats received intraperitoneal injections of α-MG (10 mg/kg) or saline every other day. Hematoxylin and eosin and Safranin-O-Fast green staining were used to evaluate the severity of cartilage lesions up to 8 weeks following surgery. α-MG inhibited the production of NO and PGE2. The elevated expression of INOS, COX-2, MMP-3, MMP-9, and MMP-13, and the degradation of Collagen II and Aggrecan, were reversed by α-MG in IL-1β-stimulated chondrocytes. In addition, IL-1β induced considerable phosphorylation of the NF-kB signaling pathway, which was inhibited by α-MG. Furthermore, the immunofluorescence staining demonstrated that α-MG could suppress IL-1β-induced p65 nuclear translocation. In vivo, cartilage treated with α-MG showed attenuated degeneration and had low Osteoarthritis Research Society International (OARSI) scores compared with the control group. Taken together, these results show that α-MG has potential therapeutic value in the treatment of OA.

Introduction

Osteoarthritis (OA) is the most common form of arthritis, causing physical limitations, disability, mental stress, and socioeconomic burden [1]. Research suggests that the disease is a breakdown of the cartilage repair process due to biomechanical and biochemical changes within the joint [2]. Oral or topical nonsteroidal anti-inflammatory drugs can relieve OA symptoms [3]. In addition, the use of hyaluronic acid, glucosamine or chondroitin can also play a role in relieving symptoms [4]. However, none of these drugs can delay the progression of OA. To date, the only effective method for the treatment of terminal OA is full joint replacement surgery. Given that few drugs can delay the progression of OA, new treatment strategies are urgently needed.

Many studies have shown that OA is related to age, obesity, trauma, and inflammation [5], [6]. Inflammatory cytokines, such as interleukin-1β (IL-1β) and tumor necrosis factor (TNF)-α are key mediators in OA pathogenesis, which is closely related to cartilage matrix degradation [7]. Matrix metalloproteinases (MMPs) is another factor closely connected with the degradation of cartilage matrix and IL-1β can regulate its expression in the cell to a certain extent [8], [39]. Hence, inhibition of inflammation may be a valid method for the treatment of OA. Recently, research on the therapeutic effects of natural compounds on human diseases has aroused much attention. For example, anti-OA compounds, such as matrine, resveratrol, and sanguinarine have been widely used in such studies [8], [9], [10].

Fruit from the tropical evergreen tree Garcinia mangostana-L (mangosteen), found in Southeast Asia, has been used in traditional therapy in the treatment of skin infections, chronic wounds and dysentery for many years [11]. α-Mangostin (α-MG), the major xanthone purified from mangosteen, has been reported to have anti-inflammatory, anti-oxidant, and anti-bacterial effects [12], [13], [14], [15]. Although there have been many studies on α-MG, there have not been reports on α-MG as a therapeutic agent for OA. Therefore, in this study, the potential beneficial effects and the underlying mechanism of α-MG on the pathogenesis of OA were investigated both in an in vitro and in vivo experimental model.

Section snippets

Reagents

α-MG (purity ≥ 98%), Collagenase type II, and dimethylsulfoxide (DMSO) were obtained from Sigma Chemical Co. (St. Louis, MO, USA). α-MG was dissolved in DMSO as a 100 mg/ml stock solution and stored at 4 °C. Further dilution was done in cell culture medium. Cell-Counting Kit-8 (CCK-8) was purchased from Dojindo (Kumamoto, Japan). Fetal bovine serum (FBS), bovine serum albumin (BSA), Dulbecco's modified Eagle's medium (DMEM)/Ham's F 12 medium and 0.25% trypsin-ethylenediaminetetraacetic acid

The potential cytotoxicity of α-MG on rat chondrocytes

The potential cytotoxicity of α-MG on rat chondrocytes was determined by the CCK-8 assay. Cells were cultured with increasing concentrations of α-MG (0, 1.25, 2.5, 5.0, and 10 μg/ml) for 24 h and 48 h, respectively. According to Fig. 1A–B, cell viabilities were not influenced by α-MG at concentrations up to 5.0 μg/ml. Results were similar between the 24 h and 48 h group. Consequently, the concentrations of α-MG (1.25, 2.5, and 5.0 μg/ml) were used in subsequent experiments.

Effect of α-MG on IL-1β-induced production of NO and PGE2 in rat chondrocytes

Studies were conducted

Discussion

OA, which is characterized by progressive joint dysfunction and pain, is the most prevalent type of chronic degenerative joint disease [1], [19]. Although there are a variety of mechanisms involved in this disease, inflammation is one of the hallmark factors [20]. There are a number of drugs available for the treatment of OA, such as steroids and nonsteroidal anti-inflammatory drugs (NSAIDs), cartilage protective agents and intra-articular injections of glucocorticoids [4], [21]. However, these

Conclusion

In summary, we demonstrated the anti-inflammatory effects of α-MG in chondrocytes. These results showed that it is possible to protect chondrocytes by inhibiting the over-expression of inflammatory mediators, and synthesis of catabolic factors probably through blocked NF-κB activation. Both in vitro and in vivo studies confirmed the protective effects of α-MG in an OA rat model as well as cell culture. However, the complete molecular mechanism of α-MG in chondrocytes has not been fully

Conflict of interest

The authors declare that they have no conflict of interest.

Reference (47)

  • J.W. Bijlsma et al.

    Osteoarthritis: an update with relevance for clinical practice

    Lancet

    (Jun 18 2011)
  • H.P. French et al.

    Prevalence and burden of osteoarthritis amongst older people in Ireland: findings from The Irish Longitudinal Study on Ageing (TILDA)

    Eur. J. Pub. Health

    (Feb 2016)
  • J.W. Bijlsma et al.

    Osteoarthritis: an update with relevance for clinical practice

    Lancet

    (Jun 18 2016)
  • L. Hauk

    Treatment of knee osteoarthritis: a clinical practice guideline from the AAOS

    Am. Fam. Physician

    (Jun 1 2014)
  • M.U. de Rezende et al.

    Drug therapy in knee osteoarthrosis

    Rev. Bras. Ortop.

    (Dec 6 2015)
  • S.A. Haq et al.

    Development of a questionnaire for identification of the risk factors for osteoarthritis of the knees in developing countries. A pilot study in Iran and Bangladesh. An ILAR-COPCORD phase III study

    Int. J. Rheum. Dis.

    (Aug 2010)
  • S. Dahaghin et al.

    Squatting, sitting on the floor, or cycling: are life-long daily activities risk factors for clinical knee osteoarthritis? Stage III results of a community-based study

    Arthritis Rheum.

    (Oct 15 2009)
  • M. Kobayashi et al.

    Role of interleukin-1 and tumor necrosis factor alpha in matrix degradation of human osteoarthritic cartilage

    Arthritis Rheum.

    (Jan 2005)
  • S. Lu et al.

    Matrine inhibits IL-1beta-induced expression of matrix metalloproteinases by suppressing the activation of MAPK and NF-kappaB in human chondrocytes in vitro

    Int. J. Clin. Exp. Pathol.

    (May 1 2015)
  • Y. Ma et al.

    Sanguinarine protects against osteoarthritis by suppressing the expression of catabolic proteases

    Oncotarget

    (Apr 11 2017)
  • D. Obolskiy et al.

    Garcinia mangostana L.: a phytochemical and pharmacological review

    Phytother. Res.

    (Aug 2009)
  • Y. Fang et al.

    Protective effect of alpha-mangostin against oxidative stress induced-retinal cell death

    Sci Rep

    (Feb 18 2016)
  • P.T. Nguyen et al.

    Antimicrobial actions of alpha-mangostin against oral streptococci

    Can. J. Microbiol.

    (Mar 2011)
  • Cited by (34)

    • Low-intensity pulsed ultrasound inhibits IL-6 in subchondral bone of temporomandibular joint osteoarthritis by suppressing the TGF-β1/Smad3 pathway

      2021, Archives of Oral Biology
      Citation Excerpt :

      Then, LIPUS was proven to inhibit the secretion of IL-1β induced inflammatory mediators through the TGF-β1/Smad3 pathway in vitro. Since IL-1β has been implicated in joint destruction as the most decisive cytokine, it has been widely used to simulate the similar microenvironment of osteoarthritis in vitro (Pan et al., 2017). Increased expression of IL-1β causes enhanced production of matrix metalloproteinases (MMPs), a disintegrin, and metalloproteinase with thrombospondin motifs (ADAMTs) and IL-6 in chondrocytes and osteoblasts (Uddin et al., 2016; Yang et al., 2020).

    • Protective effects of Erdosteine on interleukin-1β-stimulated inflammation via inhibiting the activation of MAPK, NF-κB, and Wnt/β-catenin signaling pathways in rat osteoarthritis

      2020, European Journal of Pharmacology
      Citation Excerpt :

      CCK-8 was acquired from Boster Biological Technology (Wuhan, Hubei, China). Chondrocytes were obtained from 3-day-old Sprague-Dawley rats as described in previous (Pan et al., 2017). In brief, the knee joint cartilages were isolated and cut into pieces and then digested with 0.25% trypsin-EDTA solution for 30 min and 0.2% collagenase II at 37 °C for 8 h. Then, the cell was collected and suspended in DMEM/F12 with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin antibiotics at 37°C with 5% CO2.

    View all citing articles on Scopus
    View full text