Open Access

Aminoguanidine inhibits IL‑1β‑induced protein expression of iNOS and COX‑2 by blocking the NF‑κB signaling pathway in rat articular chondrocytes

  • Authors:
    • Yuanqiang Ma
    • Xiaopeng Song
    • Tianwen Ma
    • Yue Li
    • Hui Bai
    • Zhiheng Zhang
    • Hailong Hu
    • Rui Yuan
    • Yajing Wen
    • Li Gao
  • View Affiliations

  • Published online on: July 17, 2020     https://doi.org/10.3892/etm.2020.9021
  • Pages: 2623-2630
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Osteoarthritis is a chronic joint disease which has a serious impact on the health and quality of life of affected humans and animals. As an inhibitor of inducible nitric oxide synthase (iNOS), aminoguanidine (AG) displays anti‑inflammatory effects. The purpose of the present study was to investigate the effect of AG on the expression of iNOS and cyclooxygenase‑2 (COX‑2), and the activity of the NF‑κB signaling pathway in rat chondrocytes stimulated by interleukin‑1β (IL‑1β). The viability of chondrocytes treated with AG (0.3, 1 or 3 mM) alone was determined using a Cell Counting Kit‑8 assay. Subsequently, the chondrocytes were treated with either 10 ng/ml IL‑1β alone, or co‑treated with increasing concentrations of AG (0.3, 1 or 3 mM) and 10 ng/ml IL‑1β. The protein levels of COX‑2, iNOS, phosphorylated (p)‑p65, p65, p‑NF‑κβ inhibitor α (IκBα), IκBα, p‑inhibitor of NF‑κβ‑β (IKKβ) and IKKβ were evaluated by western blotting. NF‑κB translocation was determined by immunofluorescence analysis. Western blotting and reverse transcription‑quantitative PCR were used to detect expression levels of relevant proteins/genes. The results suggested that the inhibitory effect of AG on the protein and gene expression levels of iNOS and COX‑2 in IL‑1β‑treated chondrocytes was dose‑dependent. In addition, AG decreased the level of phosphorylation of IKKβ, IκBα and NF‑κB p65, the degradation of IKKβ, IκBα and p65, and the translocation of NF‑κB in IL‑1β‑stimulated chondrocytes. The most significant inhibitory effect of AG was observed at a concentration of 1 mM. Therefore, the present study suggested that AG may serve as a potential agent to reduce the inflammatory response of chondrocytes stimulated by IL‑1β.

Introduction

Osteoarthritis (OA) is a common chronic and irreversible joint disease (1,2) that causes alterations to the morphology, structure and function of chondrocytes (3). Overexpression of inflammatory mediators, including cytokines [interleukin (IL)-1β], reactive oxygen species and matrix-degrading enzymes, leads to progressive deterioration of cartilage, synovium and subchondral bone (4). At present, no effective treatments to prevent or reverse progressive joint injury are available (5).

AG is an inducible nitric oxide synthase (iNOS) inhibitor, which can regulate the activity and expression levels of iNOS (6). iNOS inhibitors have been used to alleviate articular cartilage injury, pain and inflammation in a surgical model of OA (7). In addition, AG has various pharmacological effects, including anti-inflammatory effects that can modulate OA and articular inflammation (8). Additionally, it has been reported that AG protects against colonic inflammation by inhibiting the expression of NF-κB/p65(9). In a previous study, AG treatment reduced the protein expression levels of iNOS and p65 in the liver of an animal model of diabetes (10). Furthermore, AG can reduce the level of osteocyte apoptosis during non-traumatic osteonecrosis (11). Although the pharmacological effects of AG have been studied in numerous cell types (12-17), it remains unclear whether AG can influence iNOS and cyclooxygenase-2 (COX-2) expression, and the NF-κB signaling pathway in rat chondrocytes.

The pathogenesis of OA is related to the activation of a number of different molecular pathways, such as the NF-κB and MAPK pathways (18). Activation of the NF-κB signaling pathway directly affects the pathogenesis and development of OA (19). Activated NF-κB molecules trigger increases in the levels of other proinflammatory cytokines, for example IL-1β, tumor necrosis factor α and IL-6, leading to increased extracellular degradation and reduced synthesis of collagen and proteoglycans, which further contribute to the onset of OA (20). The activation of NF-κB has been hypothesized to be closely associated with IL-1β-stimulated OA (20). Furthermore, it has been hypothesized that the proinflammatory responses stimulated by IL-1β stimulate chondrocytes during OA, leading to increases in COX-2 and iNOS expression by activating NF-κB (21-23). In addition, NF-κB promotes articular cartilage breakdown by inducing the expression of matrix metalloproteinases and ADAMTs in OA chondrocytes (20,24). Elevated levels of NF-κB in OA chondrocytes lead to the overexpression of COX2 and iNOS, which further contribute to cartilage inflammation (25,26) and degeneration of articular cartilage (24).

Targeted therapies that interfere with NF-κB signaling may serve as a novel therapeutic strategy for the treatment of OA. In the present study, IL-1β-treated articular chondrocytes were used to investigate whether AG inhibited iNOS and COX-2 expression, and the NF-κB signaling pathway.

Materials and methods

Chondrocyte isolation and culture

Sprague-Dawley rats (age, 2-3 weeks; weight, 41±3.5 g; sex, male:female, 1:1) obtained from The Second Affiliated Hospital of Harbin Medical University (Harbin, China) were anaesthetized with isoflurane and sacrificed by cervical dislocation. Articular cartilage from the femoral heads and knees was isolated, and ground into small pieces under sterile conditions. All experiments were approved by the Departmental Animal Care and Use Committee at Northeast Agricultural University.

As previously described (27), cartilage debris was isolated with trypsin (Gibco; Thermo Fisher Scientific, Inc.) at 37˚C for 30 min, and subsequently rinsed with PBS containing penicillin and streptomycin solution. The tissue was digested with collagenase Type II (Gibco; Thermo Fisher Scientific, Inc.) in PBS at 37˚C for 4 h. Subsequently, the supernatant was collected and centrifuged at 500 x g, 27˚C for 7 min. The harvested articular chondrocytes were placed in 25 m2 culture flasks containing DMEM supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin, and were incubated at 37˚C with 5% CO2. When the monolayer on the bottom of the culture flask reached 80-90% confluence, the cells were passaged and the second-generation chondrocyte cells were isolated for subsequent experiments.

Chondrocyte treatment

To assess cell viability, chondrocytes were seeded in 96-well plates (5x104/ml, 100 µl/well), treated with 0, 0.3, 1, or 3 mM AG (Sigma-Aldrich; Merck KGaA) for 24, 48 and 72 h at 37˚C and evaluated using a Cell Counting Kit-8 (CCK-8) assay (Dojindo Molecular Technologies, Inc.), as described below. For the evaluation of iNOS and COX-2 expression, chondrocyte cells were exposed to the following conditions for 24 h at 37˚C: i) 10 ng/ml IL-1β (PeproTech, Inc.) alone or ii) co-treatment with different concentrations of AG (0.3, 1, 3 mM) and 10 ng/ml IL-1β for 24 h, after treatment with 10 ng/ml IL-1β for 24 h alone. The activity of the NF-κB signaling pathway was assessed in chondrocytes treated with either 10 ng/ml IL-1β alone for 0.5 h or co-treated with AG (0.3, 1 or 3 mM) and 10 ng/ml IL-1β at 37˚C for 2 h. Rat chondrocytes cultured without AG and IL-1β were used as controls.

Cell viability assay

The cytotoxicity of AG was determined using a CCK-8 assay. The assay was conducted on chondrocytes treated with varying concentrations of AG (0.3, 1 or 3 mM) for 24, 48, and 72 h, as aforementioned. Cells were incubated with 10 µl CCK-8 reagent at 37˚C for 2 h. Subsequently, the absorbance at a wavelength of 450 nm was determined using a microplate reader.

Western blot analysis

AG-treated chondrocyte cells were washed with PBS and collected with cell scrapers. Total protein was extracted from AG-treated chondrocyte cells using RIPA lysis buffer (Beyotime Institute of Biotechnology) by centrifugation at 10,000 x g for 15 min at 4˚C. Total protein was quantified using a bicinchoninic acid assay. Protein (15 µl/lane) was separated by 10% SDS-PAGE and transferred to nitrocellulose membranes. Subsequently, the membranes were blocked with 5% skim-milk at room temperature for 1 h. The membranes were incubated at 4˚C overnight with primary antibodies targeted against: p65 (cat. no. 8242; 1:1,000; Cell Signaling Technology, Inc.), NF-κβ inhibitor α (IκBα; cat. no. 9242; 1:1,000; Cell Signaling Technology, Inc.), inhibitor of NF-κβ-β (IKKβ; cat. no. ab124957; 1:1,000; Abcam), phosphorylated (p)-p65 (cat. no. 3033; 1:1,000; Cell Signaling Technology, Inc.), p-IκBα (cat. no. 9246; 1:1,000; Cell Signaling Technology, Inc.), p-IKKβ (cat. no. ab59195; 1:1,000; Abcam), iNOS (cat. no. WL0992a; 1:500; Wanleibio Co., Ltd.), COX-2 (cat. no. WL01750; 1:500; Wanleibio Co., Ltd.) and GAPDH (cat. no. TA-08; 1:3,000; OriGene Technologies, Inc.). Subsequently, the membranes were washed with TBST. Following primary incubation, the membranes were incubated for 2 h at 37˚C with horseradish peroxidase-conjugated secondary antibody goat anti-mouse IgG and horseradish peroxidase-conjugated secondary antibody goat anti-rabbit IgG (cat. nos. ZB2305 and ZB-2301, respectively; 1:1,000; ZSGB-BIO). Protein bands were visualized using an ECL kit (Beyotime Institute of Biotechnology) and the signals were analyzed using a Tanon detection system (Tanon Science and Technology Co., Ltd.). GAPDH was used as the loading control. The densities of bands were analyzed using ImageJ (version 1.51; National Institutes of Health).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the chondrocyte cells using the BioSci™ Tissue/Cultured Cell Total RNA Extraction kit (Dakewe Bioengineering Co., Ltd.), according to the manufacturer's instructions. Total RNA (1 µg), dNTPs (cat. no. AD101-12; Beijing TransGen Biotech Co., Ltd.), primer (cat. no. AH101-02; Beijing TransGen Biotech Co., Ltd.) and TransScript®II reverse transcriptase (cat. no. AH101-02; Beijing TransGen Biotech Co., Ltd.) were used to synthesize cDNA. The temperature protocol was as follows: 50˚C for 30 min and 85˚C for 5 sec. Quantitative PCR (qPCR) was performed using SuperReal PreMix Plus (SYBR Green; cat. no. FP205; Tiangen Biotech Co., Ltd.) according to the manufacturer's protocols. The following thermocycling conditions used for qPCR: 15 min at 95˚C for initial denaturation; followed by 40 cycles at 95˚C for 15 sec, 60˚C for 32 sec, and 72˚C for 5 min. The following primer pairs were used for qPCR: GAPDH forward, 5'-GATGCCCCCATGTTTGTGAT-3' and reverse, 5'-GGCATGGACTGTGGTCATGAG-3'; iNOS forward, 5'-GAGACGCACAGGCAGAGGTTG-3' and reverse, 5'-AGCAGGCACACGCAATGATGG-3'; and COX-2 forward, 5'-AGAAGCGAGGACCTGGGTTCA C-3' and reverse, 5'-ACACCTCTCCACCGATGACCTG-3'. Protein levels were quantified using the 2-ΔΔCq method (28) and normalized to the internal reference gene GAPDH.

Immunofluorescence assay

After treating with 10% FBS, Second-generation chondrocytes cultured with DMEM supplemented with 0.05% FBS were treated with 10 ng/ml IL-1β, or co-incubated with 10 ng/ml IL-1β and 1 mM AG for 2 h at 37˚C. Subsequently, the cells were washed with PBS and fixed with 4% paraformaldehyde for 1 h at 37˚C. The cells were washed with PBS, blocked with 10% goat serum (cat. no. AR1009; 0.3% Triton, 1:10; Boster Biological Technology) for 1 h at room temperature and rinsed with PBS. Subsequently, the cells were incubated with primary antibodies against p-p65 (cat. no. 3033; 1:100; Cell Signaling Technology, Inc.) overnight at 4˚C. Following primary incubation, the cells were gently washed and then incubated with a horseradish peroxidase-conjugated secondary antibody goat anti-rabbit IgG (cat. no. ZB-2301; 1:250; OriGene Technologies, Inc.) for 2 h at 37˚C. Subsequently, the cells were stained with DAPI (Beyotime Institute of Biotechnology) at room temperature for 15 min. Cells were rinsed and observed under a fluorescence microscope in six randomly-selected fields (magnification, x400).

Statistical analysis

Statistical analyses were performed using SPSS software (version 18.0; SPSS, Inc.). Data are presented as the mean ± standard deviation. And all experiments were performed at least three times. Differences were assessed using one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of AG on cell viability

To determine whether different concentrations of AG were cytotoxic to rat chondrocytes, cells were treated with AG at varying concentrations (0.3, 1 or 3 mM) for 24, 48, and 72 h. The results suggested that treatment with AG did not significantly alter the viability of the cells, as measured by a CCK-8 assay (P>0.05; Fig. 1).

Effect of AG on IL-1β-induced expression levels of iNOS and COX2 in chondrocytes

Chondrocytes treated with IL-1β displayed significantly increased COX-2 and iNOS expression levels compared with the control cells (Fig. 2A-E). Compared with cells stimulated with IL-1β alone, the protein expression levels of iNOS and COX-2 were decreased in a dose-dependent manner by AG (P<0.05; Fig. 2A-C). The gene expression levels of iNOS and COX-2 were also decreased in a dose-dependent manner by AG (P<0.05; Fig. 2D and E).

Effect of AG on the activity of NF-κB in chondrocytes induced by IL-1β

Western blot analysis suggested that the levels of p-IKKβ and p-IκBα were significantly increased in IL-1β-stimulated chondrocytes compared with the control cells (P<0.05; Fig. 3A, C and E). Moreover, the protein levels of p-IKKβ and p-IκBα were significantly reduced in chondrocytes co-treated with AG and IL-1β (P<0.05; Fig. 3C and E). Interestingly, the lowest p-IKKβ and p-IκBα protein expression levels were observed in chondrocytes treated with 1 mM AG (Fig. 3C and E). Chondrocyte treatment with IL-1β significantly reduced the protein expression levels of IκBα (P<0.05; Fig. 3D) and IKKβ levels (P<0.05; Fig. 3B) compared with the control cells. Chondrocytes co-cultured with AG and IL-1β displayed decreased expression levels of IKKβ and IκBα compared with the IL-1β group (Fig. 3B and D). Similar to the results obtained for p-IKKβ/IKKβ and p-IκBα/IκBα, the highest inhibitory effect of AG on IL-1β-induced p-IKKβ/IKKβ and p-IκBα/IκBα alterations was observed in cells treated with 1 mM AG (Fig. 3C and D).

Treatment with IL-1β significantly reduced the protein expression levels of NF-κB p65 in chondrocytes compared with the control cells (Fig. 3F). By contrast, co-treatment with AG and IL-1β resulted in significantly higher expression levels of p65 compared with cells stimulated with IL-1β alone (P<0.05; Fig. 3F). Compared with cells stimulated with IL-1β alone, co-treatment with 0.3, 1 or 3 mM AG and IL-1β significantly reduced the p-p65/p65 (P<0.05; Fig. 3G). Importantly, the greatest decrease in p-p65 levels was observed in the group treated with 1 mM AG (Fig. 3G). Taken together, the results suggested that AG inhibited the activity of NF-κB in chondrocytes activated by IL-1β and that AG displayed the strongest inhibitory effect at a concentration of 1 mM.

Effect of AG on the nuclear translocation of NF-κB p65

No p65 staining was observed in the nuclei of untreated control cells, indicated by no green fluorescence inside the nucleus (Fig. 4). However, IL-1β-stimulated chondrocytes displayed p65 staining in the nucleus, but not in the cytoplasm (Fig. 4). However, IL-1β-stimulated chondrocytes displayed p-p65 staining in the nucleus. Therefore, strong green fluorescence was observed in the nucleus (Fig. 4). In cells co-treated with 1 mM AG and 10 ng/ml IL-1β, the intensity of green fluorescence in the nucleus decreased. The levels of cytoplasmic staining were also low in this group (Fig. 4). Therefore, these results suggested that the entry of p-p65 into the nucleus was limited in cells co-treated with AG and IL-1β.

Discussion

OA is a chronic joint disease (1) and at present, no effective preventive or therapeutic drugs are available for the treatment of the disease (2). Therefore, the development of effective compounds for the treatment of OA is important. In the present study, the potential of AG, a compound that inhibits iNOS, to prevent or delay the progression of OA was investigated.

The roles of AG as an inhibitor of oxidation (29), apoptosis (30,31) and inflammation (32-34) have been widely recognized and accepted. In addition to these well-recognized roles, AG can inhibit the generation of isoproterenol-induced reactive oxygen species, and restore levels of antioxidant superoxide dismutase, glutathione and catalase in the heart (35). AG can also suppress the production of nitric oxide and the expression of iNOS in osteocytes, leading to the release of cytochrome C and the induction of osteocyte apoptosis (11). Moreover, AG can decrease the accumulation of glycosylation products, which can lead to endoplasmic reticulum stress-induced cell apoptosis (36). Furthermore, AG mediates articular inflammatory processes by downregulating IL-1β production in human osteoarthritic synovial tissue and cartilage (8). Therefore, in the present study, the possible associations between the inhibitory effect of AG on inflammation and the NF-κB signaling pathway were investigated.

IL-1β is involved in the pathogenesis of OA and it may result in marked alterations to the cartilage, including matrix degradation, inflammation, chondrocyte phenotypic changes and chondrocyte apoptosis (26,37-39). Previous studies have reported that 10 ng/ml IL-1β induces an inflammatory response in chondrocytes (27,40); therefore, 10 ng/ml was used as the working concentration of IL-1β in the present study. IL-1β induces iNOS and COX-2 expression, promotes the synthesis of inflammatory mediators, including prostaglandin E2 and nitric oxide, and stimulates articular chondrocytes to produce high levels of NF-κB (26). Therefore, in the present study, chondrocytes were stimulated with IL-1β to mimic the pathophysiology of OA. The results of the present study were consistent with previous studies, suggesting that IL-1β induced increased iNOS and COX-2 expression (37,41,42).

iNOS is not only an inflammatory mediator, but is also essential for the initiation and promotion of the inflammatory response (43). iNOS can significantly increase the production of nitric oxide, which leads to destruction of articular cartilage and chondrocyte apoptosis (44). In addition, iNOS can regulate other inflammatory processes, for example, cortisol can interact with or induce iNOS to increase the extent of DNA damage, and the formation of reactive oxygen/nitrogen species (45). Increased gene and protein expression levels of iNOS and COX-2 contribute to pain and joint inflammation in patients with OA (46). Moreover, current OA treatment strategies focus on the use of anti-inflammatory drugs that reduce COX-2 levels, which can alleviate joint pain and swelling (47). Therefore, reducing the expression of iNOS and COX-2 in chondrocytes could potentially ameliorate joint inflammation and the degeneration of articular cartilage in patients with OA. In a recent study, treatment with kaempferol reduced the expression levels of iNOS and COX-2 in a dose-dependent manner in IL-1β-stimulated rat chondrocytes (23). Consistently, the results of the present study indicated that AG decreased the gene and protein expression levels of iNOS and COX-2 in IL-1β-stimulated rat articular chondrocytes. Therefore, the present study suggested that AG might reduce inflammatory responses.

Furthermore, the present study suggested that the expression of COX-2 and iNOS may be closely associated with the activation of the NF-κB signaling pathway. In a study conducted by Lianxu et al (39), an small interfering RNA targeted against NF-κB p65 reduced iNOS and COX-2 expression levels in rat chondrocytes stimulated by IL-1β. Additionally, IL-1β-mediated iNOS expression was reduced following suppression of NF-κB activity in rat chondrocytes (48). The aforementioned studies suggested that increased iNOS and COX-2 protein expression levels in IL-1β-stimulated rat chondrocytes are dependent upon the activation of NF-κB. Furthermore, it has been reported that the protein expression level of IκBα and the activity of NF-κB are decreased by IKKβ phosphorylation (49). Activated NF-κB translocates into the nucleus to induce the expression of iNOS and COX-2(50) and other proinflammatory cytokines, such as NO and PGE2(26), which further aggravate OA symptoms. The results reported in the aforementioned studies were consistent with the results of the present study, which indicated that IL-1β successfully induced an inflammatory response in chondrocytes. Therefore, suppressing NF-κB activity might serve as a novel therapeutic option for OA (11,25,36,46,47). In the present study, AG inhibited IKKβ phosphorylation, IκBα degradation and NF-κB/p65 activation. Similar effects have been reported with pomegranate fruit extract (51). Pomegranate fruit extract inhibited IL-6 production, the expression of IKKβ mRNA, the degradation of IκBα and the nuclear translocation of NF-κB/p65 in OA chondrocytes. Furthermore, AG inhibited expression of COX-2 and iNOS, and similar effects have been observed with chrysin (52), suggesting that there is a common mechanism of action among these drugs. The present study suggested that AG inhibited NF-κB activation and suppressed the inflammatory response in IL-1β-stimulated chondrocytes. Therefore, it was hypothesized that AG inhibited nuclear translocation of NF-κB/p65 by inhibiting the phosphorylation of IκBα and IKKβ, thereby reducing the expression of iNOS and COX-2, and suppressing the inflammatory response. Collectively, these results indicated that AG has the potential to protect articular chondrocytes. Further investigation into the clinical application of AG is required.

In conclusion, AG downregulated iNOS and COX-2 expression by blocking the NF-κB signaling pathway; therefore, AG may protect chondrocytes. Additionally, 1 mM AG was the most effective concentration for the inhibition of inflammation. Furthermore, the present study indicated that AG may serve as a potential therapeutic for OA, therefore, providing the theoretical basis for future clinical studies.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Key R&D Program of China (grant no. 2017YFD0502200) and the Major National Science and Technology Special and Key Research Projects-Provincial Funding Projects (grant no. GX18B023).

Availability of data and materials

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

Authors' contributions

YM designed the study and prepared the manuscript. LG designed the study and produced the final draft of manuscript before submitting. YM, TM and XS analyzed the data. YM, ZZ, HB, YL, HH, RY and YW performed the experiments and processed the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Departmental Animal Care and Use Committee at Northeast Agricultural University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bijlsma JW, Berenbaum F and Lafeber FP: Osteoarthritis: An update with relevance for clinical practice. Lancet. 377:2115–2126. 2011.PubMed/NCBI View Article : Google Scholar

2 

Madry H and Cucchiarini M: Advances and challenges in gene-based approaches for osteoarthritis. J Gene Med. 15:343–355. 2013.PubMed/NCBI View Article : Google Scholar

3 

Scanzello CR and Goldring SR: The role of synovitis in osteoarthritis pathogenesis. Bone. 51:249–257. 2012.PubMed/NCBI View Article : Google Scholar

4 

Litwic AE, Parsons C, Edwards MH, Jagannath D, Cooper C and Dennison EM: Comment on: Inflammatory mediators in osteoarthritis: A critical review of the state-of-the art, prospects, and future challenges. Bone. 106:28–29. 2018.PubMed/NCBI View Article : Google Scholar

5 

McCulloch K, Litherland GJ and Rai TS: Cellular senescence in osteoarthritis pathology. Aging Cell. 16:210–218. 2017.PubMed/NCBI View Article : Google Scholar

6 

Hafez HM, Ibrahim MA, Ibrahim SA, Amin EF, Goma W and Abdelrahman AM: Potential protective effect of etanercept and aminoguanidine in methotrexate-induced hepatotoxicity and nephrotoxicity in rats. Eur J Pharmacol. 768:1–12. 2015.PubMed/NCBI View Article : Google Scholar

7 

Balaganur V, Pathak NN, Lingaraju MC, More AS, Latief N, Kumari RR, Kumar D and Tandan SK: Effect of S-methylisothiourea, an inducible nitric oxide synthase inhibitor, in joint pain and pathology in surgically induced model of osteoarthritis. Connect Tissue Res. 55:367–377. 2014.PubMed/NCBI View Article : Google Scholar

8 

Shirazi I, Yaron I, Wollman Y, Blum M, Chernihovsky T, Judovich R, Iaina A and Yaron M: Down regulation of interleukin 1beta production in human osteoarthritic synovial tissue and cartilage cultures by aminoguanidine. Ann Rheum Dis. 60:391–394. 2001.PubMed/NCBI View Article : Google Scholar

9 

Farghaly HS and Thabit RH: L-arginine and aminoguanidine reduce colonic damage of acetic acid-induced colitis in rats: Potential modulation of nuclear factor-κB/p65. ClinExp Pharmacol Physiol. 41:769–779. 2014.PubMed/NCBI View Article : Google Scholar

10 

Di Naso FC, Rodrigues G, Dias AS, Porawski M, Fillmann H and Marroni NP: Hepatic nitrosative stress in experimental diabetes. J Diabetes Complications. 26:378–381. 2012.PubMed/NCBI View Article : Google Scholar

11 

Wang J, Kalhor A, Lu S, Crawford R, Ni JD and Xiao Y: iNOS expression and osteocyte apoptosis in idiopathic, non-traumatic osteonecrosis. Acta Orthop. 86:131–141. 2015.PubMed/NCBI View Article : Google Scholar

12 

Kim J, Kim CS, Sohn E, Lee YM, Jo K, Shin SD and Kim JS: Aminoguanidine protects against apoptosis of retinal ganglion cells in Zucker diabetic fatty rats. Eur Rev Med Pharmacol Sci. 18:1573–1578. 2014.PubMed/NCBI

13 

Li W, Hu Q, Ren X, He P, Xu H, Dai H and Chen Z: Aminoguanidine suppresses methylglyoxal-mediated oxygen-glucose deprivation injury in human brain microvascular endothelial cells. Zhejiang Da Xue Xue Bao Yi Xue Ban. 42:261–266. 2013.(In Chinese). PubMed/NCBI

14 

Tian M, Qing C, Niu Y, Dong J, Cao X, Song F, Ji X and Lu S: Effect of aminoguanidine intervention on neutrophils in diabetes inflammatory cells wound healing. Exp Clin Endocrinol Diabetes. 121:635–642. 2013.PubMed/NCBI View Article : Google Scholar

15 

Sliman SM, Eubank TD, Kotha SR, Kuppusamy ML, Sherwani SI, Butler ES Kuppusamy P, Roy S, Marsh CB, Stern DM and Parinandi NL: Hyperglycemic oxoaldehyde, glyoxal, causes barrier dysfunction, cytoskeletal alterations, and inhibition of angiogenesis in vascular endothelial cells: Aminoguanidine protection. Mol Cell Biochem. 333:9–26. 2010.PubMed/NCBI View Article : Google Scholar

16 

Saiko P, Graser G, Giessrigl B, Lackner A, Grusch M, Krupitza G, Basu A, Sinha BN, Jayaprakash V, Jaeger W, et al: A novel N-hydroxy-N'-aminoguanidine derivative inhibits ribonucleotide reductase activity: Effects in human HL-60 promyelocytic leukemia cells and synergism with arabinofuranosylcytosine (Ara-C). Biochem Pharmacol. 81:50–59. 2011.PubMed/NCBI View Article : Google Scholar

17 

Zheng B, Zheng T, Wang L, Chen X, Shi C and Zhao S: Aminoguanidine inhibition of iNOS activity ameliorates cerebral vasospasm after subarachnoid hemorrhage in rabbits via restoration of dysfunctional endothelial cells. J Neurol Sci. 295:97–103. 2010.PubMed/NCBI View Article : Google Scholar

18 

Berenbaum F: Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthritis Cartilage. 21:16–21. 2013.PubMed/NCBI View Article : Google Scholar

19 

Rigoglou S and Papavassiliou AG: The NF-κB signalling pathway in osteoarthritis. Int J Biochem Cell Biol. 45:2580–2584. 2013.

20 

Wojdasiewicz P, Poniatowski ŁA and Szukiewicz D: The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm. 2014(561459)2014.PubMed/NCBI View Article : Google Scholar

21 

Liao S, Zhou K, Li D, Xie X, Jun F and Wang J: Schisantherin A suppresses interleukin-1β-induced inflammation in human chondrocytes via inhibition of NF-κB and MAPKs activation. Eur J Pharmacol. 780:65–70. 2016.PubMed/NCBI View Article : Google Scholar

22 

Rasheed N, Alghasham A and Rasheed Z: Lactoferrin from camelus dromedarius inhibits nuclear transcription factor-κB Activation, Cyclooxygenase-2 expression and prostaglandin E2 production in stimulated human chondrocytes. Pharmacognosy Res. 8:135–141. 2016.PubMed/NCBI View Article : Google Scholar

23 

Zhuang Z, Ye G and Huang B: Kaempferol alleviates the interleukin-1β-induced inflammation in rat osteoarthritis chondrocytes via suppression of NF-κB. Med Sci Monit. 23:3925–3931. 2017.PubMed/NCBI View Article : Google Scholar

24 

Chen YJ, Tsai KS, Chan DC, Lan KC, Chen CF, Yang RS and Liu SH: Honokiol, a low molecular weight natural product, prevents inflammatory response and cartilage matrix degradation in human osteoarthritis chondrocytes. J Orthop Res. 32:573–580. 2014.PubMed/NCBI View Article : Google Scholar

25 

Zhou HF, Yan H, Pan H, Hou KK, Akk A, Springer LE, Hu Y, Allen JS, Wickline SA and Pham C: Peptide-siRNA nanocomplexes targeting NF-κB subunit p65 suppress nascent experimental arthritis. J Clin Invest. 124:4363–4374. 2014.PubMed/NCBI View Article : Google Scholar

26 

Wang D, Qiao J, Zhao X, Chen T and Guan D: Thymoquinone inhibits IL-1β-induced inflammation in human osteoarthritis chondrocytes by suppressing NF-κB and MAPKs signaling pathway. Inflammation. 38:2235–2241. 2015.PubMed/NCBI View Article : Google Scholar

27 

Li Y, Wang J, Song X, Bai H, Ma T, Zhang Z, Li X, Jiang R, Wang G, Fan X, et al: Effects of baicalein on IL-1β-induced inflammation and apoptosis in rat articular chondrocytes. Oncotarget. 8:90781–90795. 2017.PubMed/NCBI View Article : Google Scholar

28 

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

29 

Ahmad R, Ahmad N, Naqvi AA, Exarchou V, Upadhyay A, Tuenter E, Foubert K, Apers S, Hermans N and Pieters L: Antioxidant and Antiglycating Constituents from Leaves of Ziziphus oxyphylla and Cedrela serrata. Antioxidants(Basel). 5(9)2016.PubMed/NCBI View Article : Google Scholar

30 

Orman D, Vardi N, Ates B, Taslidere E and Elbe H: Aminoguanidine mitigates apoptosis, testicular seminiferous tubules damage, and oxidative stress in streptozotocin-induced diabetic rats. Tissue Cell. 47:284–290. 2015.PubMed/NCBI View Article : Google Scholar

31 

Wright C, Iyer AKV, Kulkarni Y and Azad N: S-Nitrosylation of Bcl-2 negatively affects autophagy in lung epithelial cells. J Cell Biochem. 117:521–532. 2015.PubMed/NCBI View Article : Google Scholar

32 

Madhu BP, Singh KP, Saminathan M, Singh R, Tiwari AK, Manjunatha V, Harish C and Manjunathareddy GB: Correlation of inducible nitric oxide synthase (iNOS) inhibition with TNF-α, caspase-1, FasL and TLR-3 in pathogenesis of rabies in mouse model. Virus Genes. 52:61–71. 2016.PubMed/NCBI View Article : Google Scholar

33 

Shafaroodi H, Hassanipour M, Mousavi Z, Rahimi N and Dehpour AR: The effects of sub-chronic treatment with pioglitazone on the septic mice mortality in the model of cecal ligation and puncture: Involvement of nitric oxide pathway. Acta Med Iran. 53:608–616. 2015.PubMed/NCBI

34 

Tian M, Qing C, Niu Y, Dong J, Cao X, Song F, Ji X and Lu S: Aminoguanidine cream ameliorates skin tissue microenvironment in diabetic rats. Arch Med Sci. 12:179–187. 2016.PubMed/NCBI View Article : Google Scholar

35 

Parthasarathy A, Gopi V, Devi KMS, Balaji N and Vellaichamy E: Aminoguanidine inhibits ventricular fibrosis and remodeling process in isoproterenol-induced hypertrophied rat hearts by suppressing ROS and MMPs. Life Sci. 118:15–26. 2014.PubMed/NCBI View Article : Google Scholar

36 

Lenin R, Mohan V and Balasubramanyam M: SEAP activity serves for demonstrating ER stress induction by glucolipotoxicity as well as testing ER stress inhibitory potential of therapeutic agents. Mol Cell Biochem. 404:271–279. 2015.PubMed/NCBI View Article : Google Scholar

37 

Chen WP, Wang YL, Tang JL, Hu PF, Bao JP and Wu LD: Morin inhibits interleukin-1β-induced nitric oxide and prostaglandin E2 production in human chondrocytes. Int Immunopharmacol. 12:447–452. 2012.PubMed/NCBI View Article : Google Scholar

38 

Li R, Cai L, Ding J, Hu CM, Wu TN and Hu XY: Inhibition of hedgehog signal pathway by cyclopamine attenuates inflammation and articular cartilage damage in rats with adjuvant-induced arthritis. J Pharm Pharmacol. 67:963–971. 2015.PubMed/NCBI View Article : Google Scholar

39 

Lianxu C, Hongti J and Changlong Y: NF-kappaBp65-specific siRNA inhibits expression of genes of COX-2, NOS-2 and MMP-9 in rat IL-1beta-induced and TNF-alpha-induced chondrocytes. Osteoarthritis Cartilage. 14:367–376. 2006.PubMed/NCBI View Article : Google Scholar

40 

Huang X, Pan Q, Mao Z, Zhang R, Ma X, Xi Y and You H: Sinapic acid inhibits the IL-1 β-induced inflammation via MAPK downregulation in rat chondrocytes. Inflammation. 41:562–568. 2018.PubMed/NCBI View Article : Google Scholar

41 

Ma Z, Wang Y, Piao T and Liu J: Echinocystic acid inhibits IL-1β-induced COX-2 and iNOS expression in human osteoarthritis chondrocytes. Inflammation. 39:543–549. 2016.PubMed/NCBI View Article : Google Scholar

42 

Ansari MY and Haqqi TM: Interleukin-1β induced stress granules sequester COX-2 mRNA and regulates its stability and translation in human OA chondrocytes. Sci Rep. 6(27611)2016.PubMed/NCBI View Article : Google Scholar

43 

Suschek CV, Schnorr O and Kolb-Bachofen V: The role of NOS in chronic inflammatory processes in vivo: Is it damage-promoting, protective, or active at all? Curr Mol Med. 4:763–775. 2004.PubMed/NCBI View Article : Google Scholar

44 

Gómez R, Scotece M, Conde J, Lopez V, Pino J, Lago F, Gómez-Reino JJ and Gualillo O: Nitric oxide boosts TLR-4 mediated lipocalin 2 expression in chondrocytes. J Orthop Res. 31:1046–1052. 2013.PubMed/NCBI View Article : Google Scholar

45 

Flaherty RL, Owen M, Fagan-Murphy A, Intabli H, Healy D, Patel A, Allen MC, Patel BA and Flint MS: Glucocorticoids induce production of reactive oxygen species/reactive nitrogen species and DNA damage through an iNOS mediated pathway in breast cancer. Breast Cancer Res. 19(35)2017.PubMed/NCBI View Article : Google Scholar

46 

Wang SN, Xie GP, Qin CH, Chen YR, Zhang KR, Li X, Wu Q, Dong WQ, Yang J and Yu B: Aucubin prevents interleukin-1 beta induced inflammation and cartilage matrix degradation via inhibition of NF-κB signaling pathway in rat articular chondrocytes. Int Immunopharmacol. 24:408–415. 2015.PubMed/NCBI View Article : Google Scholar

47 

Chin KY: The spice for joint inflammation: Anti-inflammatory role of curcumin in treating osteoarthritis. Dove Medical Press. 10:3029–3042. 2016.PubMed/NCBI View Article : Google Scholar

48 

Lei M, Wang JG, Xiao DM, Fan M, Wang DP, Xiong JY, Chen Y, Ding Y and Liu SL: Resveratrol inhibits interleukin 1β-mediated inducible nitric oxide synthase expression in articular chondrocytes by activating SIRT1 and thereby suppressing nuclear factor-κB activity. Eur J Pharmacol. 674:73–79. 2012.PubMed/NCBI View Article : Google Scholar

49 

Ying X, Chen X, Cheng S, Shen Y, Peng L and Xu HZ: Piperine inhibits IL-β induced expression of inflammatory mediators in human osteoarthritis chondrocyte. Int Immunopharmacol. 17:293–299. 2013.PubMed/NCBI View Article : Google Scholar

50 

Stuhlmeier KM: The anti-rheumatic gold salt aurothiomalate suppresses interleukin-1beta-induced hyaluronan accumulation by blocking HAS1 transcription and by acting as a COX-2 transcriptional repressor. J Biol Chem. 282:2250–2258. 2007.PubMed/NCBI View Article : Google Scholar

51 

Haseeb A, Khan NM, Ashruf OS and Haqqi TM: A polyphenol-rich pomegranate fruit extract suppresses NF-κB and IL-6 expression by blocking the activation of IKK and NIK in primary human chondrocytes. Phytother Res. 31:778–782. 2017.PubMed/NCBI View Article : Google Scholar

52 

Zheng W, Tao Z, Cai L, Chen C, Zhang C, Wang Q, Ying X, Hu W and Chen H: Chrysin attenuates IL-1β-induced expression of inflammatory mediators by suppressing NF-κB in human osteoarthritis chondrocytes. Inflammation. 40:1143–1154. 2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma Y, Song X, Ma T, Li Y, Bai H, Zhang Z, Hu H, Yuan R, Wen Y, Gao L, Gao L, et al: Aminoguanidine inhibits IL‑1β‑induced protein expression of iNOS and COX‑2 by blocking the NF‑κB signaling pathway in rat articular chondrocytes. Exp Ther Med 20: 2623-2630, 2020
APA
Ma, Y., Song, X., Ma, T., Li, Y., Bai, H., Zhang, Z. ... Gao, L. (2020). Aminoguanidine inhibits IL‑1β‑induced protein expression of iNOS and COX‑2 by blocking the NF‑κB signaling pathway in rat articular chondrocytes. Experimental and Therapeutic Medicine, 20, 2623-2630. https://doi.org/10.3892/etm.2020.9021
MLA
Ma, Y., Song, X., Ma, T., Li, Y., Bai, H., Zhang, Z., Hu, H., Yuan, R., Wen, Y., Gao, L."Aminoguanidine inhibits IL‑1β‑induced protein expression of iNOS and COX‑2 by blocking the NF‑κB signaling pathway in rat articular chondrocytes". Experimental and Therapeutic Medicine 20.3 (2020): 2623-2630.
Chicago
Ma, Y., Song, X., Ma, T., Li, Y., Bai, H., Zhang, Z., Hu, H., Yuan, R., Wen, Y., Gao, L."Aminoguanidine inhibits IL‑1β‑induced protein expression of iNOS and COX‑2 by blocking the NF‑κB signaling pathway in rat articular chondrocytes". Experimental and Therapeutic Medicine 20, no. 3 (2020): 2623-2630. https://doi.org/10.3892/etm.2020.9021