Next Article in Journal
Hyalectanase Activities by the ADAMTS Metalloproteases
Next Article in Special Issue
Is the Membrane Lipid Matrix a Key Target for Action of Pharmacologically Active Plant Saponins?
Previous Article in Journal
Iron Availability in Tissue Microenvironment: The Key Role of Ferroportin
Previous Article in Special Issue
Maternal Reproductive Toxicity of Some Essential Oils and Their Constituents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antiosteoarthritic Effect of Morroniside in Chondrocyte Inflammation and Destabilization of Medial Meniscus-Induced Mouse Model

1
Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea
2
Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea
3
Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
4
Natural Products Research Institute, Gyeonggi Institute of Science & Technology Promotion, Suwon 16229, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(6), 2987; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22062987
Submission received: 23 February 2021 / Revised: 10 March 2021 / Accepted: 12 March 2021 / Published: 15 March 2021

Abstract

:
Osteoarthritis (OA) is a common degenerative disease that results in joint inflammation as well as pain and stiffness. A previous study has reported that Cornus officinalis (CO) extract inhibits oxidant activities and oxidative stress in RAW 264.7 cells. In the present study, we isolated bioactive compound(s) by fractionating the CO extract to elucidate its antiosteoarthritic effects. A single bioactive component, morroniside, was identified as a potential candidate. The CO extract and morroniside exhibited antiosteoarthritic effects by downregulating factors associated with cartilage degradation, including cyclooxygenase-2 (Cox-2), matrix metalloproteinase 3 (Mmp-3), and matrix metalloproteinase 13 (Mmp-13), in interleukin-1 beta (IL-1β)-induced chondrocytes. Furthermore, morroniside prevented prostaglandin E2 (PGE2) and collagenase secretion in IL-1β-induced chondrocytes. In the destabilization of the medial meniscus (DMM)-induced mouse osteoarthritic model, morroniside administration attenuated cartilage destruction by decreasing expression of inflammatory mediators, such as Cox-2, Mmp3, and Mmp13, in the articular cartilage. Transverse microcomputed tomography analysis revealed that morroniside reduced DMM-induced sclerosis in the subchondral bone plate. These findings suggest that morroniside may be a potential protective bioactive compound against OA pathogenesis.

1. Introduction

Osteoarthritis (OA) is a complex chronic disease and one of the most common types of arthritis, resulting in cartilage loss, pain, and stiffness [1,2]. The prevalence of OA is the most commonly observed among the elderly and obese, with a high risk of joint injuries [3]. Notably, the early onset of OA is initiated by mechanical stress in the articular cartilage, resulting in an excessive level of interleukin-1 beta (IL-1β) in the superficial zone of the cartilage [4,5]. Excessive levels of IL-1β trigger joint damage and inflammation via the upregulation of OA-inducing factors such as cyclooxygenase-2 (Cox-2) and matrix metalloproteinase (Mmp) [6,7]. These inflammatory mediators suppress collagen synthesis in chondrocytes following stimulation with prostaglandin E2 and collagenase secretion [8]. Recent studies have revealed that nuclear factor-κB (NF-κB) induces the expression of Cox-2, Mmp-3, and Mmp-13, while decreasing cartilage extracellular matrix (ECM) proteins, including collagen type II alpha 1 (Col2a1) synthesis [9,10]. Several pharmacological treatments against OA have focused on alleviating IL-1β mediated OA pathogenesis, including nonsteroidal anti-inflammatory drugs (NSAIDs), diacerein, and paracetamol [11,12,13]. However, these drugs have limitations with long-term administration owing to their serious side effects [14,15].
In recent years, natural plant-based treatments have been developed as alternatives for numerous diseases, with fewer side effects than conventional drugs [16,17]. Previous studies have shown that some plant extracts have preventive effects against OA pathogenesis [18,19]. Cornus officinalis (CO) of the dogwood genus has been found in Eastern Asia for more than 2000 years and has been employed as a traditional natural product for maintaining liver and kidney health [20]. Manu et al. have indicated the beneficial effects of CO on type 2 diabetes, neuroinflammation, and bone loss [21,22,23,24]. Recent studies have shown that the CO extract possesses potent anti-inflammatory action against oxidative stress in RAW 264.7 cells and decreases proinflammatory factors, such as IL-1β, interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), in numerous types of inflammation [25,26,27]. Additionally, aqueous extracts of the CO fruit reportedly inhibit OA symptoms in a testosterone-deficient mouse model [28]. Phytochemical studies have demonstrated that herbal products contain a variety of bioactive components responsible for pharmacological activities against various diseases [29,30]. Numerous pharmaceutical bioactive components isolated from the CO extract have been identified, including morroniside, loganin, ursolic acid, and caffeic acid [31].
In the present study, we aimed to identify a single bioactive compound derived from the CO extract and evaluate its anti-OA protective effects in primary chondrocyte inflammation in vitro and destabilization of the medial meniscus (DMM) mouse model in vivo.

2. Results and Discussion

2.1. Cornus Officinalis (CO) Extract Reduces Mouse Primary Chondrocyte Inflammation

Primary chondrocyte inflammation induced by IL-1β treatment has been widely employed as an experimental model for investigating OA pathogenesis [32]. Chondrocytes are localized in the articular cartilage and play an important role in the modulation of ECM homeostasis [33]. During OA development, IL-1β initiates an inflammatory signaling cascade, inducing the expression of Cox-2, Mmp-3, and Mmp-13, resulting in OA pathogenesis [34,35]. In the present study, we determined whether the CO extract possessed anti-inflammatory effects against chondrocyte inflammation. Mouse primary chondrocytes were treated with IL-1β and coincubated with different concentrations of the CO extract (2, 10, and 50 μg/mL) for 48 h. The CO extract reduced IL-1β induced Cox-2, Mmp-3, and Mmp-13 mRNA and protein expression in chondrocytes when compared with the nontreated (Mock) group (Figure 1). Furthermore, the CO extract did not affect cell viability (Supplementary Figure S1A). These results indicate that the CO extract possesses anti-inflammatory activity against chondrocyte inflammation.

2.2. Identification of Morroniside Isolated from CO Extract

The CO extract consists of several bioactive compounds with varying geological, cultivation, and climate differences [36,37]. Therefore, we performed a phytochemical screening of the CO extract fractions to elucidate the anti-inflammatory effects of the bioactive compounds on chondrocyte inflammation. High-performance liquid chromatography (HPLC) analysis of the CO extract is presented in Figure 2A. The anti-inflammatory effects of each fraction were determined by Cox-2 expression, a reliable marker for inflammatory signaling in chondrocytes [38]. Single bioactive compound(s) were fractionated by sequential fractionation (Supplementary Figure S2). Consequently, a specific bioactive compound morroniside was identified (Figure 2B) and confirmed by a combination of one- and two-dimensional nuclear magnetic resonance (NMR) spectrometry (Figure 2C,D).

2.3. Single Bioactive Compound, Morroniside, Attenuates Mouse Primary Chondrocyte Inflammation

Morroniside, an iridoid glycoside, is one of the main constituents of C. officinalis [39]. Phytochemical studies have shown that morroniside has various beneficial effects, including antidiabetic, antioxidative, cardioprotective, and antineuroinflammatory effects [40,41]. Based on previous studies, the anti-inflammatory effects of the CO extract could be exerted by morroniside. Therefore, we examined the anti-inflammatory effect of morroniside against chondrocyte inflammation. Primary cultured chondrocytes induced by IL-1β were treated with different concentrations of morroniside (2, 10, and 50 μM). In the present study, morroniside treatment attenuated inflammatory responses, such as Cox-2, Mmp-3, and Mmp-13, expression, when compared with those of the nontreated group (Figure 3). Additionally, morroniside treatment downregulated NF-κB protein levels. These results suggest that morroniside inhibits chondrocyte inflammation by reducing Cox-2, Mmp-3, Mmp-13, and NF-κB.
In OA inflammation, chondrocytes produce prostaglandin E2 (PGE2) and collagenase [42]. PGE2 plays a key role in the regulation of the inflammatory responses stimulated by Cox-2 expression [43]. In the synovial fluid, PGE2 and collagenase secretion induced by IL-1β participate in ECM degradation, resulting in mechanical stress in the subchondral region [44]. In the present study, morroniside treatment reduced IL-1β-mediated upregulation of PGE2 and collagenase levels in primary chondrocytes (Figure 4). These results indicate that morroniside prevents OA pathogenesis by reducing PGE2 and collagenase secretion.

2.4. Morroniside Administration Prevents DMM-Induced OA in Mice

The meniscus is a fibrocartilaginous tissue that extends mechanical integrity to the knee [45]. The loss of the meniscal function triggers increased mechanical stress in the articular cartilage, resulting in subchondral bone degeneration [46]. Furthermore, mechanical stress induces inflammatory responses in the articular cartilage via the secretion of IL-1β in the synovial region [47]. As the DMM mouse model mimics human OA, DMM mice have been used to investigate OA pathogenesis [48]. The DMM-induced OA model is characterized by the degeneration of the articular cartilage and osteophyte formation in response to inflammatory responses [49]. To investigate the antiosteoarthritic effect of morroniside in vivo, surgically destabilized medial meniscus mice were used to induce OA. Morroniside (5 and 20 mg/kg/day) was administered for 10 weeks. Morroniside treatment significantly reduced articular cartilage degeneration by decreasing inflammatory responses, including Cox-2, Mmp-3, and Mmp-13 expression, when compared with the nontreated DMM group (Figure 5). In addition, microcomputed tomography (micro-CT) images of the articular cartilage revealed that DMM mice presented osteophyte formation. However, morroniside inhibited DMM-induced sclerosis and cartilage degradation (Figure 6), indicating that morroniside prevents DMM-induced OA in vivo by reducing articular cartilage inflammation.
In terms of morroniside as a phytomedicine for antiosteoarthritic agents, metabolic profiles should be addressed to consider the bioavailability of the drug [50]. In the present study, we determined the bioavailability of morroniside in vivo. The blood concentrations of morroniside after a single administration were measured by HPLC analysis. Mice were orally administered morroniside (5 mg/kg) and blood was collected at 0, 5, 15, 30, 60, 120, and 240 min. According to a plasma clearance test, blood concentrations of morroniside peaked at 30 min, with almost complete removal observed within 240 min (Figure 7), indicating that morroniside was actively eliminated from the blood. These results suggest that morroniside might be a potent anti-inflammatory agent against OA pathogenesis.

3. Materials and Methods

3.1. Fractionation, Isolation, and Identification of Morroniside from CO Extract

Aerial parts of CO (1.4 kg) were dried in the shade, powdered, and added to 40 L of 70% ethanol (HPLC grade), twice at room temperature (each time for 2 days), and finally concentrated in a vacuum at 40 °C to yield 75.7 g of extract. The extracts were suspended in distilled water and Diaion HP-20 chromatography resin using gradient mixtures as eluents (100% H2O, 30% EtOH, 70% EtOH, EtOH fractions, F001–F004). Fraction F002 (30% EtOH) was separated by C18 MPLC using gradient mixtures as eluents (F011–F015). Morroniside was isolated from F012 by employing C18 MPLC. The structure was elucidated by a combination of 1D and 2D NMR spectrometry, as well as by comparison with reported literature (Figure 1) [51]. For in vitro and in vivo experiments, commercial morroniside was obtained from Sigma-Aldrich (St. Louis, MO, USA) and dissolved in distilled water.
Morroniside (1) white amorphous solid. 1H-NMR (CD3OD, 700 MHz) d: 1.31 (3H, d, J = 7.2 Hz, H-10b), 1.38 (3H, d, J = 7.2 Hz, H-10a), 1.75 (1H, ddd, J = 2.0, 4.0, 9.0 Hz, H-9b), 1.80 (1H, ddd, J = 2.0, 4.0, 9.0 Hz, H-9a), 1.88 (2H, m, H-6b), 2.00 (2H, m, H-6a), 2.82 (1H, dt, J = 4.0, 13.0 Hz, H-5a), 3.12 (1H, dt, J = 4.0, 13.0 Hz, H-5b), 3.35–3.41 (4H, m, H2, H-3, H-4, H-5), 3.70 (3H, s, OCH3), 3.87 (1H, dd, J = 2.0, 7.0 Hz, H-8a), 3.93 (1H, dd, J = 3.0, 7.0 Hz, H-8b), 4.24 (1H, dd, J = 2.0, 12.0 Hz, H6a), 4.44 (1H, dd, J = 2.0, 12.0 Hz, H-6b), 4.78 (1H, d, J = 4.0 Hz, H-7), 4.80 (1H, d, J = 8.0 Hz, H-1), 5.82 (1H, d, J = 8.6 Hz, H-1a), 5.86 (1H, d, J = 9.0 Hz, H-1b), 7.51 (1H, s, H-3);
13C-NMR (CD3OD, 175 MHz) d: 19.8 (C-10), 27.3 (C-5b), 31.9 (C-5a), 34.5 (C-6b), 37.1 (C-6a), 39.7 (C-9a), 40.4 (C-9b), 51.8 (OCH3), 62.6 (C-6), 65.8 (C-8b), 71.4 (C-2), 74.0 (C4), 74.9 (C-8a), 77.8 (C-5), 78.3 (C-3), 95.5 (C-7), 96.9 (C-1), 99.9 (C1), 110.7 (C-4), 154.4 (C-3), 168.7 (CO).

3.2. Mouse Chondrocyte Primary Culture and the Destabilization of Medial Meniscus (DMM) Mouse Model

All animal experiments in the present study were approved by the Institutional Animal Care and Use Committee of Ajou University School of Medicine (2016–0062), and experiments were conducted in accordance with the guidelines of the committee. Primary chondrocytes were isolated from the knee joint of mice as previously described [52]. Briefly, the articular cartilage of the knee joint from 5-day-old mice was dissected and incubated with a digestive solution (Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, CA, USA) containing 1% collagenase type II (Sigma), 0.5% trypsin EDTA, and 1% penicillin/streptomycin) for 2 h. Digestive suspensions were filtered through a Corning® 40 μm cell strainer (Sigma), followed by centrifugation at 1500 rpm. Cell pellets were resuspended in DMEM, supplemented with 10% fetal bovine serum (Invitrogen) and 1% penicillin/streptomycin. Next, the cells (3 × 106 cells/well) were seeded in 6-well plates and maintained at 37 °C in a humidified incubator with 5% CO2. For the DMM-induced OA mouse model, 12-week-old male C57BL/6 mice were anesthetized with tiletamine/zolazepam (Zoletil; Vibrac Laboratories, Carros, France), and the medial meniscus in the left articular cartilage was surgically removed. After surgery, mice were administered morroniside (5 mg/kg and 20 mg/kg) by oral gavage for 10 weeks. At the end of the experiment, the mice were anesthetized with Zoletil and evaluated bone mineral density (BMD) of knee joint using PIXI-mus bone densitometer with the on-board PIXI-mus software (GE Lunar, Madison, WI, USA). At the end of the experiment, the left articular cartilage was subjected to histological analysis.

3.3. Cell Viability and Cell Supernatant Analysis

Primary chondrocytes (1 × 105 cells/well) were seeded overnight in 96-well plates at 37 °C and treated with different concentrations of the CO extract (2, 10, and 50 μg/mL) or morroniside (2, 10, and 50 μM) for 48 h. Cell viability was determined using the D-Plus™ CCK cell viability assay kit (Donginbiotech, Seoul, Korea) in accordance with the manufacturer’s instructions. To analyze PGE2 and collagenase levels secreted by the chondrocytes, total cell supernatants were concentrated with Vivaspin® 2 Centrifugal Concentrator (Sartorius, Göttingen, Germany) and assessed using the Prostaglandin E2 Parameter Assay Kit (R&D Systems, Minneapolis, MN, USA) and EnzChek™ Gelatinase/Collagenase Assay Kit (Invitrogen), respectively, in accordance with the manufacturer’s instructions.

3.4. Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR) and Quantitative RT-PCR (qRT-PCR)

Total RNA from mouse primary chondrocytes was isolated using TRIzol reagent (Invitrogen) according to the manufacturer’s instructions. Complementary DNA (cDNA) was synthesized using RevertAid™ H Minus First Strand cDNA synthesis kit (Fermentas, Hanover, NH, USA), RT-PCR was performed using the HiPi Plus 5× PCR MasterMix (ELPIS Biotech, Daejeon, Korea), and the qRT-PCR was performed using SYBR Green I qPCR kit (TaKaRa, Shiga, Japan), according to the manufacturer’s instructions. The gene-specific primers for inflammatory responses in primary chondrocytes were as follows: 5′-GGT CTG GTG CCT GGT CTG ATG AT-3′ and reverse 5′-GTC CTT TCA AGG AGA ATG GTG C-3′ for mouse Cox-2; forward 5′-CTG TGT GTG GTT GTG TGC TCA TCC TAC-3′ and reverse 5′-GGC AAA TCC GGT GTA TAA TTC ACA ATC-3′ for mouse Mmp-3; forward 5′-TGA TGG ACC TTC TGG TCT GGC-3′ and reverse 5′-CAT CCA CAT GGT TGG GAA GTT CTG-3′ for mouse Mmp-13; forward 5′-TCA CTG CCA CCC AGA C-3′ and reverse 5′-TGT AGG CCA TGA GGT CCA C-3′ for mouse GAPDH. The qRT-PCR protocol was as follows: 95 °C for 10 min, followed by 40 cycles of 95 °C for 5 s, 60 °C for 30 s, and 72 °C for 30 s, and finally a terminal melting step of 72 °C to 95 °C for 2 min (5 s per degree). Gene expression was normalized to GAPDH expression. Results were determined using the 2−ΔΔCt method, and the fold change was expressed by comparison with the untreated control group.

3.5. Western Blot Analysis and Antibodies

Cells were washed thrice with phosphate-buffered saline and then lysed with radioimmunoprecipitation assay (RIPA) buffer (BIOSESANG, Seongnam, Gyeonggi-do, Korea) containing phenylmethylsulfonyl fluoride (PMSF) cocktail (Sigma) and phosphatase inhibitor cocktail (Sigma). Cell lysates were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride membranes (Merck Millipore, Burlington, MA, USA). To evaluate alterations in the levels of proteins associated with inflammatory responses, cell lysates and mouse femoral bone slides were analyzed using appropriate primary antibodies (mouse Cox-2 (sc-1745, Santa Cruz, Dallas, TX, USA), mouse Mmp-3 (ab52915, Abcam, Cambridge, UK), mouse Mmp-13 [ab51072, Abcam], NF-κB p65 (#8242, Cell Signaling Technology, Danvers, MA, USA), and β-actin (sc-47778, Santa Cruz)). The bands of three independent Western blots and IHC images were quantified by ImageJ (Version 1.53g) software (NIH, Rockville, MA, USA).

3.6. Histology and Micro-CT Analysis

Mouse knee joints were fixed with 4% paraformaldehyde for 48 h and then decalcified with 0.5 M ethylenediaminetetraacetic acid (EDTA, pH 8.0) for 2 weeks. Bone tissues were embedded in paraffin and sectioned (4 μm) using a rotary microtome. Tissue sections were stained with safranin O for assessment of the subchondral region and visualized by optical microscopy (Leica, Wetzlar, Germany). For micro-CT analysis, a paraformaldehyde-fixed femoral bone was scanned using the high-energy spiral scan micro-CT (Skyscan 1173; Bruker, Billerica, MA, USA). Two-dimensional CT images were reconstructed using the CTvox Software version 3.2 (Bruker).

3.7. Statistical Analysis

All results are presented as the mean ± standard error of the mean (SEM), and statistical analysis was performed using GraphPad Prism 5.0 (GraphPad, San Diego, CA, USA). Multiple comparisons between groups were evaluated by one-way analysis of variance (ANOVA), followed by Tukey’s honestly significant difference (HSD) post hoc test. A probability value less than 0.05 (p < 0.05) was considered statistically significant.

4. Conclusions

In the present study, we examined the antiosteoarthritic effect of a single bioactive component morroniside isolated from the CO extract. This bioactive compound was found to be responsible for the anti-inflammatory effects mediated by the CO extract. Morroniside reduced IL-1β-induced primary cultured chondrocyte inflammation by decreasing the expression of Cox-2, Mmp3, and Mmp13, as well as PGE2 and collagenase secretion. In the DMM mouse OA model, morroniside administration ameliorated cartilage degeneration, inflammatory responses, and osteophyte formation. These findings suggest that morroniside may be a putative pharmacological agent against OA.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/1422-0067/22/6/2987/s1, Figure S1: Effects of Cornus officinalis (CO) extract and morroniside on mouse primary chondrocytes; Figure S2: Fractionation and isolation of the bioactive component from CO extract.

Author Contributions

E.P., S.Y., and S.-Y.J. designed the research. C.G.L., S.H.Y., S.H., J.K., and H.J., performed the in vitro experiments and analyzed the data. C.G.L., S.J.H., S.H.Y., S.H., and H.J. performed the animal experiment. C.W.C., and S.Y. conducted fractionation and purification of the CO extract. E.P., C.G.L., S.J.H., and S.-Y.J. interpreted the data and wrote the paper. S.Y., and S.-Y.J., edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Technology development Program of the Ministry of SMEs and Startups (S2910765) and the National Research Foundation of Korea (NRF) grant funded by the Korean government (2020R1I1A1A01072353).

Institutional Review Board Statement

All animal experiments, including primary chondrocyte isolation and destabilization of the medial meniscus, were approved by the Institutional Animal Care and Use Committee of the Ajou University School of Medicine (2016–0062).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Martel-Pelletier, J.; Barr, A.J.; Cicuttini, F.M.; Conaghan, P.G.; Cooper, C.; Goldring, M.B.; Goldring, S.R.; Jones, G.; Teichtahl, A.J.; Pelletier, J.P. Osteoarthritis. Nat. Rev. Dis. Primers 2016, 2, 16072. [Google Scholar] [CrossRef] [Green Version]
  2. Di Paola, R.; Fusco, R.; Impellizzeri, D.; Cordaro, M.; Britti, D.; Morittu, V.M.; Evangelista, M.; Cuzzocrea, S. Adelmidrol, in combination with hyaluronic acid, displays increased anti-inflammatory and analgesic effects against monosodium iodoacetate-induced osteoarthritis in rats. Arthritis Res. Ther. 2016, 18, 291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sacitharan, P.K. Ageing and osteoarthritis. Subcell. Biochem. 2019, 91, 123–159. [Google Scholar] [PubMed]
  4. Rim, Y.A.; Ju, J.H. The role of fibrosis in osteoarthritis progression. Life 2020, 11, 3. [Google Scholar] [CrossRef]
  5. Impellizzeri, D.; Siracusa, R.; Cordaro, M.; Peritore, A.F.; Gugliandolo, E.; D’Amico, R.; Fusco, R.; Crupi, R.; Rizzarelli, E.; Cuzzocrea, S.; et al. Protective effect of a new hyaluronic acid-carnosine conjugate on the modulation of the inflammatory response in mice subjected to collagen-induced arthritis. Biomed. Pharmacother. 2020, 125, 110023. [Google Scholar] [CrossRef]
  6. Aida, Y.; Maeno, M.; Suzuki, N.; Shiratsuchi, H.; Motohashi, M.; Matsumura, H. The effect of IL-1β on the expression of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases in human chondrocytes. Life Sci. 2005, 77, 3210–3221. [Google Scholar] [CrossRef]
  7. Philipot, D.; Guerit, D.; Platano, D.; Chuchana, P.; Olivotto, E.; Espinoza, F.; Dorandeu, A.; Pers, Y.M.; Piette, J.; Borzi, R.M.; et al. p16INK4a and its regulator miR-24 link senescence and chondrocyte terminal differentiation-associated matrix remodeling in osteoarthritis. Arthritis Res. Ther. 2014, 16, R58. [Google Scholar] [CrossRef] [Green Version]
  8. Fei, J.; Liang, B.; Jiang, C.; Ni, H.; Wang, L. Luteolin inhibits IL-1β-induced inflammation in rat chondrocytes and attenuates osteoarthritis progression in a rat model. Biomed. Pharmacother. 2019, 109, 1586–1592. [Google Scholar] [CrossRef]
  9. Jimi, E.; Fei, H.; Nakatomi, C. NF-κB signaling regulates physiological and pathological chondrogenesis. Int. J. Mol. Sci. 2019, 20, 6275. [Google Scholar] [CrossRef] [Green Version]
  10. Park, C.; Jeong, J.W.; Lee, D.S.; Yim, M.J.; Lee, J.M.; Han, M.H.; Kim, S.; Kim, H.S.; Kim, G.Y.; Park, E.K.; et al. Sargassum serratifolium extract attenuates interleukin-1β-induced oxidative stress and inflammatory response in chondrocytes by suppressing the activation of NF-κB, p38 MAPK, and PI3K/Akt. Int. J. Mol. Sci. 2018, 19, 2308. [Google Scholar] [CrossRef] [Green Version]
  11. Hermann, W.; Lambova, S.; Muller-Ladner, U. Current treatment options for osteoarthritis. Curr. Rheumatol. Rev. 2018, 14, 108–116. [Google Scholar] [CrossRef]
  12. Onakpoya, I.J. Paracetamol as first line for treatment of knee and hip osteoarthritis. BMJ Evid. Based Med. 2020, 25, 40. [Google Scholar] [CrossRef]
  13. Lindler, B.N.; Long, K.E.; Taylor, N.A.; Lei, W. Use of herbal medications for treatment of osteoarthritis and rheumatoid arthritis. Medicines 2020, 7, 67. [Google Scholar] [CrossRef]
  14. Patel, J.; Ladani, A.; Sambamoorthi, N.; LeMasters, T.; Dwibedi, N.; Sambamoorthi, U. A machine learning approach to identify predictors of potentially inappropriate non-steroidal anti-inflammatory drugs (NSAIDs) use in older adults with osteoarthritis. Int. J. Environ. Res. Public Health 2021, 18, 155. [Google Scholar] [CrossRef]
  15. McCrae, J.C.; Morrison, E.E.; MacIntyre, I.M.; Dear, J.W.; Webb, D.J. Long-term adverse effects of paracetamol—A review. Br. J. Clin. Pharmacol. 2018, 84, 2218–2230. [Google Scholar] [CrossRef] [Green Version]
  16. Tran, N.; Pham, B.; Le, L. Bioactive compounds in anti-diabetic plants: From herbal medicine to modern drug discovery. Biology 2020, 9, 952. [Google Scholar] [CrossRef]
  17. Britti, D.; Crupi, R.; Impellizzeri, D.; Gugliandolo, E.; Fusco, R.; Schievano, C.; Morittu, V.M.; Evangelista, M.; Di Paola, R.; Cuzzocrea, S. A novel composite formulation of palmitoylethanolamide and quercetin decreases inflammation and relieves pain in inflammatory and osteoarthritic pain models. BMC Vet. Res. 2017, 13, 229. [Google Scholar] [CrossRef]
  18. Fusco, R.; Siracusa, R.; Peritore, A.F.; Gugliandolo, E.; Genovese, T.; D’Amico, R.; Cordaro, M.; Crupi, R.; Mandalari, G.; Impellizzeri, D.; et al. The role of cashew (Anacardium occidentale L.) nuts on an experimental model of painful degenerative joint disease. Antioxidants 2020, 9, 511. [Google Scholar] [CrossRef] [PubMed]
  19. Gugliandolo, E.; Peritore, A.F.; Impellizzeri, D.; Cordaro, M.; Siracusa, R.; Fusco, R.; D’Amico, R.; Di Paola, R.; Schievano, C.; Cuzzocrea, S.; et al. Dietary supplementation with palmitoyl-glucosamine co-micronized with curcumin relieves osteoarthritis pain and benefits joint mobility. Animals 2020, 10, 1827. [Google Scholar] [CrossRef]
  20. Liu, X.; Han, Z.P.; Wang, Y.L.; Gao, Y.; Zhang, Z.Q. Analysis of the interactions of multicomponents in Cornus officinalis Sieb. et Zucc. with human serum albumin using on-line dialysis coupled with HPLC. J. Chromatogr. B 2011, 879, 599–604. [Google Scholar] [CrossRef]
  21. Tian, W.; Zhao, J.; Lee, J.H.; Akanda, M.R.; Cho, J.H.; Kim, S.K.; Choi, Y.J.; Park, B.Y. Neuroprotective effects of Cornus officinalis on stress-induced hippocampal deficits in rats and H2O2-induced neurotoxicity in SH-SY5Y neuroblastoma cells. Antioxidants 2019, 9, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ji, L.L.; Wang, X.; Li, J.J.; Zhong, X.J.; Zhang, B.; Juan, J.; Shang, X.Y. New iridoid derivatives from the fruits of Cornus officinalis and their neuroprotective activities. Molecules 2019, 24, 625. [Google Scholar] [CrossRef] [Green Version]
  23. Han, Y.; Jung, H.W.; Park, Y.K. Selective therapeutic effect of Cornus officinalis fruits on the damage of different organs in STZ-induced diabetic rats. Am. J. Chin. Med. 2014, 42, 1169–1182. [Google Scholar] [CrossRef]
  24. Kim, J.Y.; Kim, Y.K.; Choi, M.K.; Oh, J.; Kwak, H.B.; Kim, J.J. Effect of Cornus officinalis on receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation. J. Bone Metab. 2012, 19, 121–127. [Google Scholar] [CrossRef] [Green Version]
  25. Lee, H.-J.; Lim, H.-J.; Kim, D.-H.; Sim, B.-Y.; Lim, M.-H. Anti-Inflammatory effect of Cornus officinalis fruit extract for potential of cosmetic ingredient. Kor. J. Aesthet. Cosmetol. 2015, 13, 461–468. [Google Scholar]
  26. Hwang, K.A.; Hwang, Y.J.; Song, J. Antioxidant activities and oxidative stress inhibitory effects of ethanol extracts from Cornus officinalis on raw 264.7 cells. BMC Complement. Altern. Med. 2016, 16, 1–9. [Google Scholar] [CrossRef] [Green Version]
  27. Quah, Y.; Lee, S.J.; Lee, E.B.; Birhanu, B.T.; Ali, M.S.; Abbas, M.A.; Boby, N.; Im, Z.E.; Park, S.C. Cornus officinalis ethanolic extract with potential anti-allergic, anti-inflammatory, and antioxidant activities. Nutrients 2020, 12, 3317. [Google Scholar] [CrossRef]
  28. Park, S.; Moon, B.R.; Kim, J.E.; Kim, H.J.; Zhang, T. Aqueous extracts of Morus alba root Bark and Cornus officinalis fruit protect against osteoarthritis symptoms in testosterone-deficient and osteoarthritis-induced rats. Pharmaceutics 2020, 12, 1245. [Google Scholar] [CrossRef] [PubMed]
  29. Zhao, S.; Baik, O.D.; Choi, Y.J.; Kim, S.M. Pretreatments for the efficient extraction of bioactive compounds from plant-based biomaterials. Crit. Rev. Food Sci. Nutr. 2014, 54, 1283–1297. [Google Scholar] [CrossRef]
  30. Sasidharan, S.; Chen, Y.; Saravanan, D.; Sundram, K.M.; Latha, L.Y. Extraction, isolation and characterization of bioactive compounds from plants’ extracts. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Ma, W.; Wang, K.-J.; Cheng, C.-S.; Yan, G.-Q.; Lu, W.-L.; Ge, J.-F.; Cheng, Y.-X.; Li, N. Bioactive compounds from Cornus officinalis fruits and their effects on diabetic nephropathy. J. Ethnopharmacol. 2014, 153, 840–845. [Google Scholar] [CrossRef]
  32. Nummenmaa, E.; Hämäläinen, M.; Pemmari, A.; Moilanen, L.J.; Tuure, L.; Nieminen, R.M.; Moilanen, T.; Vuolteenaho, K.; Moilanen, E. Transient receptor potential ankyrin 1 (TRPA1) is involved in upregulating interleukin-6 expression in osteoarthritic chondrocyte models. Int. J. Mol. Sci. 2020, 22, 87. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, J.; Verdonk, P.; Elewaut, D.; Veys, E.M.; Verbruggen, G. Homeostasis of the extracellular matrix of normal and osteoarthritic human articular cartilage chondrocytes in vitro. Osteoarthr. Cartil. 2003, 11, 801–809. [Google Scholar] [CrossRef] [Green Version]
  34. Goldring, M.B.; Otero, M.; Plumb, D.A.; Dragomir, C.; Favero, M.; El Hachem, K.; Hashimoto, K.; Roach, H.I.; Olivotto, E.; Borzì, R.M.; et al. Roles of inflammatory and anabolic cytokines in cartilage metabolism: Signals and multiple effectors converge upon MMP-13 regulation in osteoarthritis. Eur. Cells Mater. 2011, 21, 202–220. [Google Scholar] [CrossRef]
  35. Ahmad, R.; Sylvester, J.; Zafarullah, M. MyD88, IRAK1 and TRAF6 knockdown in human chondrocytes inhibits interleukin-1-induced matrix metalloproteinase-13 gene expression and promoter activity by impairing MAP kinase activation. Cell. Signal. 2007, 19, 2549–2557. [Google Scholar] [CrossRef] [PubMed]
  36. Jung, Y.-A.; Jung, Y.-S.; Hwang, G.-S. 1H NMR-based metabolomic study of Cornus officinalis from different geographical origin. J. Korean Magn. Reson. Soc. 2011, 15, 90–103. [Google Scholar] [CrossRef]
  37. Czerwińska, M.E.; Melzig, M.F. Cornus mas and Cornus officinalis—Analogies and differences of two medicinal plants traditionally used. Front. Pharmacol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  38. Timur, U.T.; Caron, M.M.J.; Jeuken, R.M.; Bastiaansen-Jenniskens, Y.M.; Welting, T.J.M.; van Rhijn, L.W.; van Osch, G.; Emans, P.J. Chondroprotective actions of selective COX-2 inhibitors in vivo: A systematic review. Int. J. Mol. Sci. 2020, 21, 6962. [Google Scholar] [CrossRef] [PubMed]
  39. Xu, M.; Wu, H.Y.; Liu, H.; Gong, N.; Wang, Y.R.; Wang, Y.X. Morroniside, a secoiridoid glycoside from Cornus officinalis, attenuates neuropathic pain by activation of spinal glucagon-like peptide-1 receptors. Br. J. Pharmacol. 2017, 174, 580–590. [Google Scholar] [CrossRef] [Green Version]
  40. Yokozawa, T.; Yamabe, N.; Kim, H.Y.; Kang, K.S.; Hur, J.M.; Park, C.H.; Tanaka, T. Protective effects of morroniside isolated from Corni fructus against renal damage in streptozotocin-induced diabetic rats. Biol. Pharm. Bull. 2008, 31, 1422–1428. [Google Scholar] [CrossRef] [Green Version]
  41. Pi, W.X.; Feng, X.P.; Ye, L.H.; Cai, B.C. Combination of morroniside and diosgenin prevents high glucose-induced cardiomyocytes apoptosis. Molecules 2017, 22, 163. [Google Scholar] [CrossRef] [Green Version]
  42. Liu, C.C.; Zhang, Y.; Dai, B.L.; Ma, Y.J.; Zhang, Q.; Wang, Y.; Yang, H. Chlorogenic acid prevents inflammatory responses in IL-1β stimulated human SW1353 chondrocytes, a model for osteoarthritis. Mol. Med. Rep. 2017, 16, 1369–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Fan, H.W.; Liu, G.Y.; Zhao, C.F.; Li, X.F.; Yang, X.Y. Differential expression of COX-2 in osteoarthritis and rheumatoid arthritis. Genet. Mol. Res. 2015, 14, 12872–12879. [Google Scholar] [CrossRef] [PubMed]
  44. Attur, M.; Al-Mussawir, H.E.; Patel, J.; Kitay, A.; Dave, M.; Palmer, G.; Pillinger, M.H.; Abramson, S.B. Prostaglandin E2 exerts catabolic effects in osteoarthritis cartilage: Evidence for signaling via the EP4 receptor. J. Immunol. 2008, 181, 5082–5088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Makris, E.A.; Hadidi, P.; Athanasiou, K.A. The knee meniscus: Structure-function, pathophysiology, current repair techniques, and prospects for regeneration. Biomaterials 2011, 32, 7411–7431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Choi, M.C.; Choi, W.H. Mithramycin A alleviates osteoarthritic cartilage destruction by inhibiting HIF-2α expression. Int. J. Mol. Sci. 2018, 19, 1411. [Google Scholar] [CrossRef] [Green Version]
  47. Bondeson, J.; Blom, A.B.; Wainwright, S.; Hughes, C.; Caterson, B.; van den Berg, W.B. The role of synovial macrophages and macrophage-produced mediators in driving inflammatory and destructive responses in osteoarthritis. Arthritis Rheum. 2010, 62, 647–657. [Google Scholar] [CrossRef] [Green Version]
  48. Culley, K.L.; Dragomir, C.L.; Chang, J.; Wondimu, E.B.; Coico, J.; Plumb, D.A.; Otero, M.; Goldring, M.B. Mouse models of osteoarthritis: Surgical model of posttraumatic osteoarthritis induced by destabilization of the medial meniscus. Methods Mol. Biol. 2015, 1226, 143–173. [Google Scholar]
  49. Utomo, L.; Eijgenraam, S.M.; Meuffels, D.E.; Bierma-Zeinstra, S.M.A.; Bastiaansen-Jenniskens, Y.M.; van Osch, G. Meniscal extrusion and degeneration during the course of osteoarthritis in the murine collagenase-induced osteoarthritis model. J. Orthop. Res. 2018, 36, 2416–2420. [Google Scholar] [CrossRef]
  50. Batiha, G.E.; Olatunde, A.; El-Mleeh, A.; Hetta, H.F.; Al-Rejaie, S.; Alghamdi, S.; Zahoor, M.; Beshbishy, A.M.; Murata, T.; Zaragoza-Bastida, A.; et al. Bioactive compounds, pharmacological actions, and pharmacokinetics of wormwood (Artemisia absinthium). Antibiotics 2020, 9, 353. [Google Scholar] [CrossRef]
  51. Sunghwa, F.; Sakurai, H.; Saiki, I.; Koketsu, M. Iodine-catalyzed etherification of morroniside. Chem. Pharm. Bull. 2009, 57, 112–115. [Google Scholar] [CrossRef] [Green Version]
  52. Cho, C.; Kang, L.J.; Jang, D.; Jeon, J.; Lee, H.; Choi, S.; Han, S.J.; Oh, E.; Nam, J.; Kim, C.S.; et al. Cirsium japonicum var. maackii and apigenin block Hif-2α-induced osteoarthritic cartilage destruction. J. Cell. Mol. Med. 2019, 23. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effects of Cornus officinalis (CO) extract on chondrocyte inflammation. Mouse primary chondrocytes were incubated with interleukin 1 beta (IL-1β, 1 ng/mL) and cotreated with the CO extract (2, 10, and 50 μg/mL) for 48 h. (A) Mouse mRNA expressions of Cox-2, Mmp-3, and Mmp-13 were examined by reverse-transcriptase polymerase chain reaction (RT-PCR, left) and quantitative RT-PCR (qRT-PCR, right). Glyceraldehyde 3-phosphate dehydrogenase (Gapdh) was used as an internal control. (B) Western blot analysis of Cox-2, Mmp-3, and Mmp-13 proteins in mouse primary chondrocytes. GAPDH was used as an internal control. The representative results are shown in the left panel, with quantitative data presented as a graph in the right panel. The results of triplicate experiments were examined by one-way analysis of variance (ANOVA, Tukey’s honestly significant difference post hoc test, analysis of variance). * p < 0.05 vs. Mock, # p < 0.05 vs. CO (2). Con; control, Mock; nontreated, CO; Cornus officinalis extract; IL-1β, interleukin-1 beta; Cox-2, cyclooxygenase-2 MMP-3, metalloproteinase 3; MMP-13, metalloproteinase 13; qRT-PCR; quantitative reverse transcriptase-polymerase chain reaction.
Figure 1. Effects of Cornus officinalis (CO) extract on chondrocyte inflammation. Mouse primary chondrocytes were incubated with interleukin 1 beta (IL-1β, 1 ng/mL) and cotreated with the CO extract (2, 10, and 50 μg/mL) for 48 h. (A) Mouse mRNA expressions of Cox-2, Mmp-3, and Mmp-13 were examined by reverse-transcriptase polymerase chain reaction (RT-PCR, left) and quantitative RT-PCR (qRT-PCR, right). Glyceraldehyde 3-phosphate dehydrogenase (Gapdh) was used as an internal control. (B) Western blot analysis of Cox-2, Mmp-3, and Mmp-13 proteins in mouse primary chondrocytes. GAPDH was used as an internal control. The representative results are shown in the left panel, with quantitative data presented as a graph in the right panel. The results of triplicate experiments were examined by one-way analysis of variance (ANOVA, Tukey’s honestly significant difference post hoc test, analysis of variance). * p < 0.05 vs. Mock, # p < 0.05 vs. CO (2). Con; control, Mock; nontreated, CO; Cornus officinalis extract; IL-1β, interleukin-1 beta; Cox-2, cyclooxygenase-2 MMP-3, metalloproteinase 3; MMP-13, metalloproteinase 13; qRT-PCR; quantitative reverse transcriptase-polymerase chain reaction.
Ijms 22 02987 g001
Figure 2. HPLC chromatogram of (A) total CO extract and (B) standard morroniside compound and (C) 1D and (D) 2D NMR spectrometry. HPLC, high-performance liquid chromatography; CO, Cornus officinalis; NMR, nuclear magnetic resonance.
Figure 2. HPLC chromatogram of (A) total CO extract and (B) standard morroniside compound and (C) 1D and (D) 2D NMR spectrometry. HPLC, high-performance liquid chromatography; CO, Cornus officinalis; NMR, nuclear magnetic resonance.
Ijms 22 02987 g002
Figure 3. Effects of morroniside on chondrocyte inflammation. Mouse primary chondrocytes were incubated with IL-1β (1 ng/mL) and cotreated with morroniside (2, 10, and 50 μM) for 48 h. (A) mRNA expression levels of mouse Cox-2, Mmp-3, and Mmp-13 were examined by RT-PCR (left) and qRT-PCR (right). (B) Protein levels of Cox-2, Mmp-3, Mmp-13, and NF-κB were analyzed by Western blotting. GAPDH was used as an internal control. The representative results are shown in the left panel, with quantitative data presented as a graph in the right panel. The results of triplicate experiments were examined by one-way ANOVA (Tukey’s honestly significant difference post hoc test, analysis of variance). * p < 0.05 vs. Mock, # p < 0.05 vs. MO (2). Con; control, Mock; nontreated, MO; morroniside; IL-1β, interleukin-1 beta; Cox-2, cyclooxygenase-2; MMP-3, metalloproteinase 3; MMP-13, metalloproteinase 13; qRT-PCR; quantitative reverse transcriptase-polymerase chain reaction.
Figure 3. Effects of morroniside on chondrocyte inflammation. Mouse primary chondrocytes were incubated with IL-1β (1 ng/mL) and cotreated with morroniside (2, 10, and 50 μM) for 48 h. (A) mRNA expression levels of mouse Cox-2, Mmp-3, and Mmp-13 were examined by RT-PCR (left) and qRT-PCR (right). (B) Protein levels of Cox-2, Mmp-3, Mmp-13, and NF-κB were analyzed by Western blotting. GAPDH was used as an internal control. The representative results are shown in the left panel, with quantitative data presented as a graph in the right panel. The results of triplicate experiments were examined by one-way ANOVA (Tukey’s honestly significant difference post hoc test, analysis of variance). * p < 0.05 vs. Mock, # p < 0.05 vs. MO (2). Con; control, Mock; nontreated, MO; morroniside; IL-1β, interleukin-1 beta; Cox-2, cyclooxygenase-2; MMP-3, metalloproteinase 3; MMP-13, metalloproteinase 13; qRT-PCR; quantitative reverse transcriptase-polymerase chain reaction.
Ijms 22 02987 g003
Figure 4. Effects of morroniside on prostaglandin E2 (PGE2) and collagenase secretion in IL-1β induced mouse primary chondrocytes. Cells were incubated with IL-1β (1 ng/mL) and cotreated with morroniside (2, 10, and 50 μM) for 48 h. PGE2 and collagenase levels in the total cell supernatant were determined. The results of triplicate experiments were evaluated by one-way ANOVA (Tukey’s honestly significant difference post hoc test). * p < 0.05 vs. Mock, # p < 0.05 vs. MO (2). Con; control, Mock; nontreated; MO, morroniside; IL-1β, interleukin-1 beta.
Figure 4. Effects of morroniside on prostaglandin E2 (PGE2) and collagenase secretion in IL-1β induced mouse primary chondrocytes. Cells were incubated with IL-1β (1 ng/mL) and cotreated with morroniside (2, 10, and 50 μM) for 48 h. PGE2 and collagenase levels in the total cell supernatant were determined. The results of triplicate experiments were evaluated by one-way ANOVA (Tukey’s honestly significant difference post hoc test). * p < 0.05 vs. Mock, # p < 0.05 vs. MO (2). Con; control, Mock; nontreated; MO, morroniside; IL-1β, interleukin-1 beta.
Ijms 22 02987 g004
Figure 5. Effects of morroniside on DMM-induced OA pathogenesis in mice. Mice were treated with morroniside (5 and 20 mg/kg/day) for 10 weeks. (A) Articular cartilage sections were stained with safranin O and IHC for Cox-2, Mmp-3, and Mmp-13 was performed. Representative images were visualized by light microscopy. Red circle indicates the degeneration of the articular cartilage. (B) IHC images were quantified and determined using one-way ANOVA (Tukey’s honest significant difference post hoc test, analysis of variance). * p < 0.05 vs. DMM. # p < 0.05 vs. DMM+MO 5 mg. DMM, destabilization of the medial meniscus; OA, osteoarthritis; IHC, immunohistochemistry; Sham; sham-operated, MO; morroniside; Cox-2, cyclooxygenase-2; MMP-3, metalloproteinase 3; MMP-13, metalloproteinase 13. Scale bars: 50–250 μm.
Figure 5. Effects of morroniside on DMM-induced OA pathogenesis in mice. Mice were treated with morroniside (5 and 20 mg/kg/day) for 10 weeks. (A) Articular cartilage sections were stained with safranin O and IHC for Cox-2, Mmp-3, and Mmp-13 was performed. Representative images were visualized by light microscopy. Red circle indicates the degeneration of the articular cartilage. (B) IHC images were quantified and determined using one-way ANOVA (Tukey’s honest significant difference post hoc test, analysis of variance). * p < 0.05 vs. DMM. # p < 0.05 vs. DMM+MO 5 mg. DMM, destabilization of the medial meniscus; OA, osteoarthritis; IHC, immunohistochemistry; Sham; sham-operated, MO; morroniside; Cox-2, cyclooxygenase-2; MMP-3, metalloproteinase 3; MMP-13, metalloproteinase 13. Scale bars: 50–250 μm.
Ijms 22 02987 g005
Figure 6. Effect of morroniside administration on DMM-induced osteophyte development. DMM-induced OA mice were treated with morroniside (5 and 20 mg/kg/day) for 10 weeks. (A) Representative micro-CT images of mouse knee joints reveal the density of the chondral bone plate. Red circle indicates osteophyte formation in response to inflammatory responses. (B) Bone mineral density (BMD) of proximal tibial epiphysis was analyzed using one-way ANOVA (Tukey’s honest significant difference post hoc test, analysis of variance). * p < 0.05 vs. DMM, DMM, destabilization of the medial meniscus; OA, osteoarthritis; CT, computed tomography; Sham; sham-operated, MO; morroniside.
Figure 6. Effect of morroniside administration on DMM-induced osteophyte development. DMM-induced OA mice were treated with morroniside (5 and 20 mg/kg/day) for 10 weeks. (A) Representative micro-CT images of mouse knee joints reveal the density of the chondral bone plate. Red circle indicates osteophyte formation in response to inflammatory responses. (B) Bone mineral density (BMD) of proximal tibial epiphysis was analyzed using one-way ANOVA (Tukey’s honest significant difference post hoc test, analysis of variance). * p < 0.05 vs. DMM, DMM, destabilization of the medial meniscus; OA, osteoarthritis; CT, computed tomography; Sham; sham-operated, MO; morroniside.
Ijms 22 02987 g006
Figure 7. Metabolomic profile of morroniside in mice. Mean plasma concentration–time curves of morroniside in mice after a single administration (5 mg/kg). The data are presented mean ± standard error of the mean (SEM) (n = 3).
Figure 7. Metabolomic profile of morroniside in mice. Mean plasma concentration–time curves of morroniside in mice after a single administration (5 mg/kg). The data are presented mean ± standard error of the mean (SEM) (n = 3).
Ijms 22 02987 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Park, E.; Lee, C.G.; Han, S.J.; Yun, S.H.; Hwang, S.; Jeon, H.; Kim, J.; Choi, C.W.; Yang, S.; Jeong, S.-Y. Antiosteoarthritic Effect of Morroniside in Chondrocyte Inflammation and Destabilization of Medial Meniscus-Induced Mouse Model. Int. J. Mol. Sci. 2021, 22, 2987. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22062987

AMA Style

Park E, Lee CG, Han SJ, Yun SH, Hwang S, Jeon H, Kim J, Choi CW, Yang S, Jeong S-Y. Antiosteoarthritic Effect of Morroniside in Chondrocyte Inflammation and Destabilization of Medial Meniscus-Induced Mouse Model. International Journal of Molecular Sciences. 2021; 22(6):2987. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22062987

Chicago/Turabian Style

Park, Eunkuk, Chang Gun Lee, Seong Jae Han, Seung Hee Yun, Seokjin Hwang, Hyoju Jeon, Jeonghyun Kim, Chun Whan Choi, Siyoung Yang, and Seon-Yong Jeong. 2021. "Antiosteoarthritic Effect of Morroniside in Chondrocyte Inflammation and Destabilization of Medial Meniscus-Induced Mouse Model" International Journal of Molecular Sciences 22, no. 6: 2987. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22062987

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop