• Title/Summary/Keyword: nuclear factor κB

Search Result 375, Processing Time 0.023 seconds

Baicalin suppresses lipopolysaccharide-induced matrix metalloproteinase expression: action via the mitogen-activated protein kinase and nuclear factor κB-related protein signaling pathway

  • Ko, Seon-Yle
    • International Journal of Oral Biology
    • /
    • v.46 no.1
    • /
    • pp.51-59
    • /
    • 2021
  • Periodontal disease is an inflammatory disease that affects the destruction of the bone supporting the tooth and connective tissues surrounding it. Periodontal ligament fibroblasts (PDLFs) induce overexpression of matrix metalloproteinase (MMP) involved in periodontal disease's inflammatory destruction. Osteoclasts take part in physiological bone remodeling, but they are also involved in bone destruction in many kinds of bone diseases, including osteoporosis and periodontal disease. This study examined the effect of baicalin on proteolytic enzymes' production and secretion of inflammatory cytokines in PDLFs and RAW 264.7 cells under the lipopolysaccharide (LPS)-induced inflammatory conditions. Baicalin inhibited the expression of the protein, MMP-1 and MMP-2, without affecting PDLFs' cell viability, suggesting its possibility because of the inhibition of phosphorylation activation of mitogen-activated protein kinase's p38, and the signal transduction process of nuclear factor κB (NFκB)-related protein. Also, baicalin reduced the expression of MMP-8 and MMP-9 in RAW 264.7 cells. This reduction is thought to be due to the inhibition of the signal transduction process of NFκB-related proteins affected by inhibiting p65RelA phosphorylation. Also, baicalin inhibited the secretion of nitric oxide and interleukin-6 induced by LPS in RAW 264.7 cells. These results suggest that baicalin inhibits connective tissue destruction in periodontal disease. The inhibition of periodontal tissue destruction may be a therapeutic strategy for treating inflammatory periodontal-diseased patients.

Curcumin Inhibits Osteoclastogenesis by Decreasing Receptor Activator of Nuclear Factor-κB Ligand (RANKL) in Bone Marrow Stromal Cells

  • Oh, Sora;Kyung, Tae-Wook;Choi, Hye-Seon
    • Molecules and Cells
    • /
    • v.26 no.5
    • /
    • pp.486-489
    • /
    • 2008
  • Curcumin (diferuloylmethane), a pigment derived from turmeric, has anti-oxidant and anti-inflammatory activities. Accumulating evidence points to a biochemical link between increased oxidative stress and reduced bone density. Osteoclast formation was evaluated in co-cultures of bone marrow stromal cells (BMSC) and whole bone marrow cells (BMC). Expression of receptor activator of nuclear factor-${\kappa}B$ ligand (RANKL) was analyzed at the mRNA and protein levels. Exposure to curcumin led to dose-dependent suppression of osteoclastogenesis in the co-culture system, and to reduced expression of RANKL in $IL-1{\alpha}$-stimulated BMSCs. Addition of RANKL abolished the inhibition of osteoclastogenesis by curcumin, whereas the addition of prostaglandin $E_2$ ($PGE_2$) did not. The decreased osteoclastogenesis induced by curcumin may reduce bone loss and be of potential benefit in preventing and/or attenuating osteoporosis.

Exosome-mediated lnc-ABCA12-3 promotes proliferation and glycolysis but inhibits apoptosis by regulating the toll-like receptor 4/nuclear factor kappa-B signaling pathway in esophageal squamous cell carcinoma

  • Junliang Ma;Yijun Luo;Yingjie Liu;Cheng Chen;Anping Chen;Lubiao Liang;Wenxiang Wang;Yongxiang Song
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.27 no.1
    • /
    • pp.61-73
    • /
    • 2023
  • Esophageal squamous cell carcinoma (ESCC) is a kind of malignant tumor with high incidence and mortality in the digestive system. The aim of this study is to explore the function of lnc-ABCA12-3 in the development of ESCC and its unique mechanisms. RT-PCR was applied to detect gene transcription levels in tissues or cell lines like TE-1, EC9706, and HEEC cells. Western blot was conducted to identify protein expression levels of mitochondrial apoptosis and toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling pathway. CCK-8 and EdU assays were carried out to measure cell proliferation, and cell apoptosis was examined by flow cytometry. ELISA was used for checking the changes in glycolysis-related indicators. Lnc-ABCA12-3 was highly expressed in ESCC tissues and cells, which preferred it to be a candidate target. The TE-1 and EC9706 cells proliferation and glycolysis were obviously inhibited with the downregulation of lnc-ABCA12-3, while apoptosis was promoted. TLR4 activator could largely reverse the apoptosis acceleration and relieved the proliferation and glycolysis suppression caused by lnc-ABCA12-3 downregulation. Moreover, the effect of lnc-ABCA12-3 on ESCC cells was actualized by activating the TLR4/NF-κB signaling pathway under the mediation of exosome. Taken together, the lnc-ABCA12-3 could promote the proliferation and glycolysis of ESCC, while repressing its apoptosis probably by regulating the TLR4/NF-κB signaling pathway under the mediation of exosome.

Effect of saccharin on inflammation in 3T3-L1 adipocytes and the related mechanism

  • Kim, Hye Lin;Ha, Ae Wha;Kim, Woo Kyoung
    • Nutrition Research and Practice
    • /
    • v.14 no.2
    • /
    • pp.109-116
    • /
    • 2020
  • BACKGROUND/OBJECTIVES: Excessive intake of simple sugars induces obesity and increases the risk of inflammation. Thus, interest in alternative sweeteners as a sugar substitute is increasing. The purpose of this study was to determine the effect of saccharin on inflammation in 3T3-L1 adipocytes. MATERIALS/METHODS: 3T3-L1 preadipocytes were differentiated into adipocytes. The adipocytes were treated with saccharin (0, 50, 100, and 200 ㎍/mL) for 24 h. Inflammation was induced by exposure of treated adipocytes to lipopolysaccharide (LPS) for 18 h and cell proliferation was measured. The concentration of nitric oxide (NO) was measured by using Griess reagent. Protein expressions of nuclear factor kappa B (NF-κB) and inhibitor κB (IκB) were determined by western blot analysis. The mRNA expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin 1β (IL-1β), interleukin 6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and tumor necrosis factor-α (TNF-α) were determined by real-time PCR. RESULTS: Compared with the control group, the amount of NO and the mRNA expression of iNOS in the LPS-treated group were increased by about 17.6% and 46.9%, respectively, (P < 0.05), and those parameter levels were significantly decreased by saccharin treatment (P < 0.05). Protein expression of NF-κB was decreased and that of IκB was increased by saccharin treatment (P < 0.05). Saccharin decreased the mRNA expression of COX-2 and the inflammation cytokines (IL-1β, IL-6, MCP-1, and TNF-α) (P < 0.05). CONCLUSIONS: The results of this study suggest that saccharin can inhibit LPS-induced inflammatory responses in 3T3-L1 adipocytes via the NF-κB pathway.

Antioxidant and Anti-inflammatory Effects of Extracts from the Flowers of Weigela subsessilis on RAW 264.7 Macrophages (RAW 264.7 대식세포에 미치는 병꽃나무 꽃 추출물의 항산화 및 항염증 효과)

  • Yoo, Yung Choon;Lee, Gye Won;Cho, Young Ho
    • Journal of Life Science
    • /
    • v.26 no.3
    • /
    • pp.338-345
    • /
    • 2016
  • This study investigated the antioxidant and anti-inflammatory activity of ethanol extract from the flowers of Weigela subsessilis (WS-E) in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. The total polyphenol and flavonoid content was 719.19±0.04 μg tannic acid equivalents/ml and 644.87±0.02 μg quercetin equivalents/ml, respectively. The antioxidant activities of WS-E were measured by 1,1-diphenyl-2-picrylhydrazyl (DPPH) and superoxide anion radical scavenging activity. The antioxidant activities of WS-E increased markedly, in a dose-dependent manner. To screen for anti-inflammatory agents, the inhibitory effects of WS-E on the production of proinflammatory cytokines in the LPS-stimulated RAW 264.7 macrophages was examined. WS-E had no effect on cell viability at a concentration of 100 μg/ml. Nitric oxide (NO) and interleukin (IL)-6 production were inhibited in a dose-dependent manner (p<0.05). WS-E had no effect on the production of tumor necrosis factor (TNF)-α at a concentration of 0.16–20 μg/ml but induced TNF-α at a concentration of 100 μg/ml. Inducible nitric oxide synthase (iNOS) expression was also inhibited at lower concentrations (p<0.05). In addition, WS-E reduced the activation of nuclear factor (NF)-κB by inhibition of inhibitoy (I) κB phosphorylation in RAW 264.7 macrophages upon stimulation with LPS (100 ng/ml) for 24 h but not that of mitogen-activated protein kinase (MAPK). These results suggest that WS-E may be a useful antioxidant and anti-inflammatory agent in functional cosmetics.

Anti-inflammatory effect of Sihosogan-tang via inhibition of NF-κB and MAPK cascade (NF-κB와 MAPK억제를 통한 시호소간탕(柴胡疏肝湯)의 항염증효과)

  • Hyo Jeong Jin;Sang Mi Park;Ye Lim Kim;Sung Hui Byun;Sang Chan Kim
    • Herbal Formula Science
    • /
    • v.31 no.2
    • /
    • pp.99-109
    • /
    • 2023
  • Objectives : Sihosogan-tang (SST) is one of the traditional herbal formula and also one of the Korean medical insurance medicines. It commonly used in the treatment of hepatitis, chronic gastritis, intercostal neuralgia, pleurisy, and depression in East Asia. In the present study, we have demonstrated the anti-inflammatory effects of SST in macrophage cell line. Methods : To investigate mechanism of the anti-inflammatory effect of SST, we examined the productions of nitric oxide (NO) and pro-inflammatory cytokines, and the expressions of inducible NO synthase (iNOS), nuclear factor-κ B (NF-κB) and mitogen-activated protein kinase (MAPK) on RAW 264.7 cells activated by LPS. Results : SST significantly inhibited the expression of iNOS increased by LPS, and also significantly inhibited the production of NO. In addition, SST significantly inhibited pro-inflammatory cytokines such as TNF- α and interleukines. SST inhibited the expression of NF-κB and MAPK activation. Conclusions : These results suggest that SST ameliorates inflammatory response in LPS-activated RAW 264.7 cells through the inhibition of the NF-κB and MAPK pathway. Therefore, this study supplies objective evidence for the anti-inflammatory effect of SST.

Efonidipine Inhibits JNK and NF-κB Pathway to Attenuate Inflammation and Cell Migration Induced by Lipopolysaccharide in Microglial Cells

  • Nguyen, Ngoc Minh;Duong, Men Thi Hoai;Nguyen, Phuong Linh;Bui, Bich Phuong;Ahn, Hee-Chul;Cho, Jungsook
    • Biomolecules & Therapeutics
    • /
    • v.30 no.5
    • /
    • pp.455-464
    • /
    • 2022
  • Efonidipine, a calcium channel blocker, is widely used for the treatment of hypertension and cardiovascular diseases. In our preliminary study using structure-based virtual screening, efonidipine was identified as a potential inhibitor of c-Jun N-terminal kinase 3 (JNK3). Although its antihypertensive effect is widely known, the role of efonidipine in the central nervous system has remained elusive. The present study investigated the effects of efonidipine on the inflammation and cell migration induced by lipopolysaccharide (LPS) using murine BV2 and human HMC3 microglial cell lines and elucidated signaling molecules mediating its effects. We found that the phosphorylations of JNK and its downstream molecule c-Jun in LPS-treated BV2 cells were declined by efonidipine, confirming the finding from virtual screening. In addition, efonidipine inhibited the LPS-induced production of pro-inflammatory factors, including interleukin-1β (IL-1β) and nitric oxide. Similarly, the IL-1β production in LPS-treated HMC3 cells was also inhibited by efonidipine. Efonidipine markedly impeded cell migration stimulated by LPS in both cells. Furthermore, it inhibited the phosphorylation of inhibitor kappa B, thereby suppressing nuclear translocation of nuclear factor-κB (NF-κB) in LPS-treated BV2 cells. Taken together, efonidipine exerts anti-inflammatory and anti-migratory effects in LPS-treated microglial cells through inhibition of the JNK/NF-κB pathway. These findings imply that efonidipine may be a potential candidate for drug repositioning, with beneficial impacts on brain disorders associated with neuroinflammation.

Antineuroinflammatory Effects of 7,3',4'-Trihydroxyisoflavone in Lipopolysaccharide-Stimulated BV2 Microglial Cells through MAPK and NF-κB Signaling Suppression

  • Kim, Seon-Kyung;Ko, Yong-Hyun;Lee, Youyoung;Lee, Seok-Yong;Jang, Choon-Gon
    • Biomolecules & Therapeutics
    • /
    • v.29 no.2
    • /
    • pp.127-134
    • /
    • 2021
  • Neuroinflammation―a common pathological feature of neurodegenerative disorders such as Alzheimer's disease―is mediated by microglial activation. Thus, inhibiting microglial activation is vital for treating various neurological disorders. 7,3',4'-Trihydroxyisoflavone (THIF)―a secondary metabolite of the soybean compound daidzein―possesses antioxidant and anticancer properties. However, the effects of 7,3',4'-THIF on microglial activation have not been explored. In this study, antineuroinflammatory effects of 7,3',4'-THIF in lipopolysaccharide (LPS)-stimulated BV2 microglial cells were examined. 7,3',4'-THIF significantly suppressed the production of the proinflammatory mediators nitric oxide (NO), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) as well as of the proinflammatory cytokine interleukin-6 (IL-6) in LPS-stimulated BV2 microglial cells. Moreover, 7,3',4'-THIF markedly inhibited reactive oxygen species (ROS) generation. Western blotting revealed that 7,3',4'-THIF diminished LPS-induced phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), glycogen synthase kinase-3β (GSK-3β), and nuclear factor kappa B (NF-κB). Overall, 7,3',4'-THIF exerts antineuroinflammatory effects against LPS-induced microglial activation by suppressing mitogen-activated protein kinase (MAPK) and NF-κB signaling, ultimately reducing proinflammatory responses. Therefore, these antineuroinflammatory effects of 7,3',4'-THIF suggest its potential as a therapeutic agent for neurodegenerative disorders.

Paeoniflorin ameliorates Aβ-stimulated neuroinflammation via regulation of NF-κB signaling pathway and Aβ degradation in C6 glial cells

  • Cho, Eun Ju;Kim, Hyun Young;Lee, Ah Young
    • Nutrition Research and Practice
    • /
    • v.14 no.6
    • /
    • pp.593-605
    • /
    • 2020
  • BACKGROUND/OBJECTIVES: Alzheimer's disease is common age-related neurodegenerative condition characterized by amyloid beta (Aβ) accumulation that leads cognitive impairment. In the present study, we investigated the protective effect of paeoniflorin (PF) against Aβ-induced neuroinflammation and the underlying mechanism in C6 glial cells. MATERIALS/METHODS: C6 glial cells were treated with PF and Aβ25-35, and cell viability, nitric oxide (NO) production, and pro-inflammatory cytokine release were measured. Furthermore, the mechanism underlying the effect of PF on inflammatory responses and Aβ degradation was determined by Western blot. RESULTS: Aβ25-35 significantly reduced cell viability, but this reduction was prevented by the pretreatment with PF. In addition, PF significantly inhibited Aβ25-35-induced NO production in C6 glial cells. The secretion of interleukin (IL)-6, IL-1β, and tumor necrosis factor-alpha was also significantly reduced by PF. Further mechanistic studies indicated that PF suppressed the production of these pro-inflammatory cytokines by regulating the nuclear factor-kappa B (NF-κB) pathway. The protein levels of inducible NO synthase and cyclooxygenase-2 were downregulated and phosphorylation of NF-κB was blocked by PF. However, PF elevated the protein expression of inhibitor kappa B-alpha and those of Aβ degrading enzymes, insulin degrading enzyme and neprilysin. CONCLUSIONS: These findings indicate that PF exerts protective effects against Aβ-mediated neuroinflammation by inhibiting NF-κB signaling, and these effects were associated with the enhanced activity of Aβ degradation enzymes.

Role of Nuclear Factor (NF)-κB Activation in Tumor Growth and Metastasis (종양의 성장 및 전이에 있어서 NF-κB의 역할)

  • Ko, Hyun-Mi;Choi, Jung-Hwa;Ra, Myung-Suk;Im, Suhn-Young
    • IMMUNE NETWORK
    • /
    • v.3 no.1
    • /
    • pp.38-46
    • /
    • 2003
  • Background: Platelet-activating factor (PAF) induces nuclear factor $(NF)-{\kappa}B$ activation and angiogenesis and increases tumor growth and pulmonary tumor metastasis in vivo. The role of $NF-{\kappa}B$ activation in PAF-induced angiogenesis in a mouse model of Matrigel implantation, and in PAF-mediated pulmonary tumor metastasis were investigated. Methods: Angiogenesis using Matrigel and experimental pulmonary tumor metastasis were tested in a mouse model. Electrophoretic mobility shift assay was done for the assessment of $NF-{\kappa}B$ translocation to the nucleus. Expression of angiogenic factors, such as tumor necrosis factor $(TNF)-{\alpha}$, interleukin $(IL)-1{\alpha}$, basic fibroblast growth factor (bFGF), and vascular endothelial growth factor (VEGF) were tested by RT-PCR and ELISA. Results: PAF induced a dose- and time-dependent angiogenic response. PAF-induced angiogenesis was significantly blocked by PAF antagonist, CV6209, and inhibitors of $NF-{\kappa}B$ expression or action, including antisense oligonucleotides to p65 subunit of $NF-{\kappa}B$ (p65 AS) and antioxidants such as ${\alpha}$-tocopherol and N-acetyl-L-cysteine. In vitro, PAF activated the transcription factor, $NF-{\kappa}B$ and induced mRNA expression of $TNF-{\alpha}$, $IL-1{\alpha}$, bFGF, VEGF, and its receptor, KDR. The PAF-induced expression of the above mentioned factors was inhibited by p65 AS or antioxidants. Also, protein synthesis of VEGF was increased by PAF and inhibited by p65 AS or antioxidants. The angiogenic effect of PAF was blocked when anti-VEGF antibodies was treated or antibodies against $TNF-{\alpha}$, $IL-1{\alpha}$, and bFGF was co-administrated, but not by antibodies against $TNF-{\alpha}$, $IL-1{\alpha}$, and bFGF each alone. PAF-augmented pulmonary tumor metastasis was inhibited by p65 AS or antioxidants. Conclusion: These data indicate that PAF increases angiogenesis and pulmonary tumor metastasis through $NF-{\kappa}B$ activation and expression of $NF-{\kappa}B$-dependent angiogenic factors.