• Title/Summary/Keyword: NF-${\kappa}B$/MAPK

Search Result 291, Processing Time 0.02 seconds

Rodgersia podophylla Leaves Suppress Inflammatory mediators through activation of Nrf2/HO-1 signaling, and inhibition of LPS-induced NF-κB and MAPKs signaling in RAW264.7 cells

  • Kim, Ha Na;Kim, Jeong Dong;Park, Su Bin;Jeong, Jin Boo
    • Proceedings of the Plant Resources Society of Korea Conference
    • /
    • 2019.04a
    • /
    • pp.94-94
    • /
    • 2019
  • In this study, we elucidated the anti-inflammatory mechanisms of leaves extracts from Rodgersia podophylla (RPL) in RAW264.7 cells. RP-L significantly inhibited the production of the proinflammatory mediators such as NO, iNOS, IL-$1{\beta}$ and IL-6 in LPS-stimulated RAW264.7 cells. RPL increased HO-1 expression in RAW264.7 cells, and the inhibition of HO-1 by ZnPP reduced the inhibitory effect of RPL against LPS-induced NO production in RAW264.7 cells. Inhibition of p38, ROS and $GSK3{\beta}$ attenuated RPL-mediated HO-1 expression. Inhibition of ROS inhibited p38 phosphorylation and $GSK3{\beta}$ expression induced by RPL. In addition, inhibition of $GSK3{\beta}$ blocked RPL-mediated p38 phosphorylation. RPL induced nuclear accumulation of Nrf2, and Inhibition of p38, ROS and $GSK3{\beta}$ abolished RPL-mediated nuclear accumulation of Nrf2. Furthermore, RPL blocked LPS-induced degradation of $I{\kappa}B-{\alpha}$ and nuclear accumulation of p65. RP-L also attenuated LPS-induced phosphorylation of ERK1/2 and p38. Our results suggest that RPL exerts potential antiinflammatory activity by activating ROS/$GSK3{\beta}$/p38/Nrf2/HO-1 signaling and inhibiting NF-${\kappa}B$ and MAPK signaling in RAW264.7 cells. These findings suggest that RPL may have great potential for the development of anti-inflammatory drug.

  • PDF

Anti-Inflammatory Activity of Dichloromethane Fraction from Katsuwonus pelamis Heart in LPS-Induced RAW 264.7 Cells and Mouse Ear Edema (Lipopolysaccharide로 자극된 RAW 264.7 세포와 마우스 귀부종 모델에 대한 참치 심장 Dichloromethane 분획물의 항염증 효과)

  • Kim, Min-Ji;Bae, Nan-Young;Choi, Hyeun-Deok;Kim, Koth-Bong-Woo-Ri;Park, Sun-Hee;Sung, Nak-Yun;Byun, Eui-Hong;Nam, Hee-Sup;Ahn, Dong-Hyun
    • Microbiology and Biotechnology Letters
    • /
    • v.45 no.2
    • /
    • pp.101-109
    • /
    • 2017
  • This study investigated the effect of the dichloromethane fraction form Katsuwonus pelamis heart on anti-inflammatory responses in lipopolysaccharide-stimulated RAW 264.7 cells and mouse models. Ethanol extract was partitioned with dichloromethane, ethyl acetate, butanol, and water. Among the fractions, the dichloromethane fraction showed a significant decrease in nitric oxide (NO) and pro-inflammatory cytokines [interleukin (IL)-6, $IL-1{\beta}$, and tumor necrosis $factor-{\alpha}$] production compared to ethanol extract. The dichloromethane fraction attenuated the expression of inducible nitric oxide synthase and nuclear $factor-{\kappa}B$ ($NF-{\kappa}B$) p65 proteins in a dose-dependent manner. In addition, the expression of phosphorylation of mitogen-activated protein kinases (MAPKs) was also inhibited by the dichloromethane fraction. Moreover, the administration of 10, 50, and 250 mg/kg body weight-dose dependently inhibited the formation of edema by croton-oil and the application of dichloromethane (2 mg/ear) significantly reduced epidermal and dermal thickness and the infiltrated mast cell numbers. Therefore, the dichloromethane fraction exhibited an anti-inflammation effect by inhibiting $NF-{\kappa}B$ and MAPK signaling activation in macrophages.

Effect of the Flavonoid Luteolin for Dextran Sodium Sulfate-induced Colitis in NF-${\kappa}B^{EGFP}$ Transgenic Mice (Dextran Sodium Sulfate 유발 장염 모델에서 루테올린의 치료효과)

  • Jang, Byung-Ik
    • Journal of Yeungnam Medical Science
    • /
    • v.23 no.1
    • /
    • pp.26-35
    • /
    • 2006
  • Background: Luteolin, a flavone found in various Chinese herbal medicines is known to possess anti-inflammatory properties through its ability to inhibit various proinflammatory signaling pathways including NF-${\kappa}B$ and p38 MAPK. In this study, we investigated the potential therapeutic effect of luteolin on dextran sodium sulfate (DSS)-induced colitis. Materials and Methods: We used a transgenic mouse model expressing the enhanced green fluorescent protein (EGFP) under the transcriptional control of NF-${\kappa}B$ $cis$-elements. C57BL/6 NF-${\kappa}B^{EGFP}$ mice received 2.5% DSS in their drinking water for six days in combination with daily luteolin administration (1mg/kg body weight, 0.1ml vol, intragastric) or vehicle. NF-${\kappa}B$ activity was assessed macroscopically with a Charge-Coupled Device (CCD) camera and microscopically by confocal analysis. Results: A significant increase in the Disease Activity Index (DAI), histological score (p<0.05), IL-12 p40 secretion in colonic stripe culture (p<0.05) and EGFP expression was observed in luteolin and/or DSS-treated mice compared to water-treated mice. Interestingly, a trend toward a worse colitis (DAI, IL-12p40) was observed in luteolin-treated mice compared to non-treated DSS-exposed mice. In addition, EGFP expression (NF-${\kappa}B$ activity) strongly increased in the luteolin-treated mice compared to control mice. Confocal microscopy showed that EGFP positive cells were primarily lamina propria immune cells. Conclusions: These results suggest that luteolin is not a therapeutic alternative for intestinal inflammatory disorders derived for primary defects in barrier function. Thus, therapeutic intervention targeting these signaling pathways should be viewed with caution.

  • PDF

Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Activates Pro-Survival Signaling Pathways, Nuclear Factor-${\kappa}B$ and Extracellular Signal-Regulated Kinase 1/2 in Trophoblast Cell Line, JEG-3

  • Ka Hakhyun
    • Reproductive and Developmental Biology
    • /
    • v.29 no.2
    • /
    • pp.101-108
    • /
    • 2005
  • Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) is a well-known inducer of apoptotic cell death in many tumor cells. 1RAIL is expressed in human placenta, and cytotrophoblast cells express 1RAIL receptors. However, the role of TRAIL in human placentas and cytotrophoblast cells is not. well understood. In this study a trophoblast cell line, JEG-3, was used as a model system to examine the effect of TRAIL. on key intracellular signaling pathways involved in the control of trophoblastic cell apoptosis and survival JEG-3 cells expressed receptors for 1RAIL, death receptor (DR) 4, DR5, decoy receptor (OcR) 1 and DeR2. Recombinant human TRAIL (rhTRAIL) did not have a cytotoxic effect determined by MIT assay and did not induce apoptotic cell death determined by poly-(ADP-ribose) polymerase cleavage assay. rhTRAIL induced a rapid and transient nuclear translocation of nuclear $factor-{\kappa}B(NF-{\kappa}B)$ determined by immunoblotting using nuclear protein extracts. rhTRAIL rapidly activated extracellular signal-regulated protein kinase (ERK) 1/2 as determined by immnoblotting for phospho-ERK1/2. However, c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (p38MAPK) and Akt (protein kinase B) were not activated by rhTRAIL. The ability of 1RAIL to induce $NF-{\kappa}B$ and ERK1/2 suggests that interaction between TRAIL and its receptors may play an important role in trophoblast cell function during pregnancy.

Effects of Esculetin on TNF-α Induced MMP-1 Expression in Human Fibroblasts Hs68 (인간섬유아세포 Hs68에서 esculetin이 TNF-α로 유도된 MMP-1 발현에 미치는 효과)

  • Bo Hee Jeon;Yong Min Kim
    • Korean Journal of Pharmacognosy
    • /
    • v.54 no.1
    • /
    • pp.1-8
    • /
    • 2023
  • The skin is an important barrier that protects the body from harmful environments such as UV rays. When the skin is repeatedly stimulated, such as UV rays, ROS and pro-inflammatory cytokines are overproduced. As a result, the proteins and nucleic acids that make up the skin are damaged, and aging occurs. Esculetin is known to have anti-inflammatory, antioxidant and UV-induced MMP-1 inhibitory effects. However, the inhibitory effect of MMP-1 on TNF-α-induced fibroblasts is not known. Therefore, in this study, the MMP-1 inhibitory effect of esculetin was confirmed in TNF-α-induced fibroblasts. As a result of confirming the cytotoxicity of esculetin in Hs68 cells by MTT assay, there was no significant toxicity up to 200 µM. As a result of real-time PCR and ELISA, it was confirmed that esculetin inhibited the expression of MMP-1. Esculetin did not inhibit MAPK (ERK, JNK, p38) phosphorylation, but inhibited phosphorylation of the mTOR-p70S6k signaling pathway. In addition, it was confirmed that the phosphorylation of the transcription factor NF-κB was inhibited. These results suggest that esculetin has potential as an anti-aging material.

Uncaria sinensis(OIi.) Havil Suppressendotoxin-induced Proinflammatory Responses through Blocking ERK Signaling Pathways

  • Park, Tai-Guang;Moon, Hyung-Cheal;Cho, Hae-Joong;Seo, Sang-Wan;Jung, Won-Sek;Song, Ho-Joon;Park, Sung-Joo
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.21 no.5
    • /
    • pp.1297-1302
    • /
    • 2007
  • Uncaria sinensis(OIi.) Havil (USH) is used in traditional Korean medicine to treat inflammation such as amebic dysentery. In this study, we investigated the anti-inflammatory effect of USH. The water extract of USH significantly inhibits lipopolysaccharide (LPS)-induced nitrite oxide (NO), tumor necrosis factor $(TNF)-{\alpha}$, interleukin (IL)-6and IL-12 productions in murine peritoneal macrophages. Furthermore, USH selectively inhibited activation of the extracellular signal-regulated kinase (ERK) but not of p38 MAPK, c-Jun N-terminal kinase (JNK) and nuclear $factor-{\kappa}B$ $(NF-{\kappa}B)$. In murine model, we found that administration of USH reduced serum levels of $TNF-{\alpha}$, IL-6 and IL-12 productions in LPS-treated mice. Our results suggest that USH exerts ant-inflammatory effects in macrophages via inhibition of ERK activation and may be a useful therapeutic approach to inflammatory diseases.

Vaccinium oldhamii Stems Inhibit Pro-inflammatory Response and Osteoclastogenesis through Inhibition of NF-κB and MAPK/ATF2 Signaling Activation in LPS-stimulated RAW264.7 Cells

  • Park, Su Bin;Kim, Ha Na;Kim, Jeong Dong;Jeong, Jin Boo
    • Proceedings of the Plant Resources Society of Korea Conference
    • /
    • 2019.10a
    • /
    • pp.67-67
    • /
    • 2019
  • Vaccinium oldhamii (V. oldhamii) has been reported to exert a variety of the pharmacological properties such as anti-oxidant activity, anti-cancer activity, and inhibitory activity of ${\alpha}$-amylase and acetylcholinesterase. However, the anti-inflammatory activity of V. oldhamii has not been studied. In this study, we aimed to investigate anti-inflammatory activity of the stem extracts from V. oldhamii, and to elucidate the potential mechanisms in LPS-stimulated RAW264.7 cells. Among VOS, VOL and VOF, the inhibitory effect of NO and PGE2 production induced by LPS was highest in VOS treatment. Thus, VOS was selected for the further study. VOS dose-dependently blocked LPS-induced NO and PGE2 production by inhibiting iNOS and COX-2 expression, respectively. VOS inhibited the expression of pro-inflammatory cytokines such as $IL-1{\beta}$, IL-6 and $TNF-{\alpha}$. In addition, VOS suppressed TRAP activity and attenuated the expression of the osteoclast-specific genes such as NFATc1, c-FOS, TRAP, MMP-9, cathepsin K, CA2, OSCAR and ATPv06d2. VOS inhibited LPS-induced $NF-{\kappa}B$ signaling activation through blocking $I{\kappa}B-{\alpha}$ degradation and p65 nuclear accumulation. VOS inhibited MAPK signaling activation by attenuating the phosphorylation of ERK1/2, p38 and JNK. Furthermore, VOS inhibited ATF2 phosphorylation and blocked ATF2 nuclear accumulation. From these findings, VOS has potential to be a candidate for the development of chemopreventive or therapeutic agents for the inflammatory diseases.

  • PDF

Oxidative Stress, Chromatin Remodeling and Gene Transcription in Inflammation and Chronic Lung Diseases

  • Rahman, Irfan
    • BMB Reports
    • /
    • v.36 no.1
    • /
    • pp.95-109
    • /
    • 2003
  • Inflammatory lung diseases are characterized by chronic inflammation and oxidant/antioxidant imbalance. The sources of the increased oxidative stress in patients with chronic inflammatory lung diseases such as asthma and chronic obstructive pulmonary disease (COPD) derive from the increased burden of inhaled oxidants, and from the increased amounts of reactive oxygen species (ROS) generated by several inflammatory, immune and various structural cells of the airways. Increased levels of ROS produced in the airways is reflected by increased markers of oxidative stress in the airspaces, sputum, breath, lungs and blood in patients with lung diseases. ROS, either directly or via the formation of lipid peroxidation products such as 4-hydroxy-2-nonenal may play a role in enhancing the inflammation through the activation of stress kinases (JNK, MAPK, p38) and redox sensitive transcription factors such as NF-${\kappa}B$ and AP-1. Recent evidences have indicated that oxidative stress and pro-inflammatory mediators can alter nuclear histone acetylation/deacetylation allowing access for transcription factor DNA binding leading to enhanced pro-inflammatory gene expression in various lung cells. Understanding of the mechanisms of redox signaling, NF-${\kappa}B$/AP-1 regulation, the balance between histone acetylation and deacetylation and the release and expression of pro- and anti-inflammatory mediators may lead to the development of novel therapies based on the pharmacological manipulation of antioxidants in lung inflammation and injury. Antioxidants that have effective wide spectrum activity and good bioavailability, thiols or molecules which have dual antioxidant and anti-inflammatory activity, may be potential therapeutic agents which not only protect against the direct injurious effects of oxidants, but may fundamentally alter the underlying inflammatory processes which play an important role in the pathogenesis of chronic inflammatory lung diseases.

JS-III-49, a hydroquinone derivative, exerts anti-inflammatory activity by targeting Akt and p38

  • Yi, Young-Su;Kim, Mi-Yeon;Cho, Jae Youl
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.21 no.3
    • /
    • pp.345-352
    • /
    • 2017
  • Since previous studies have reported that hydroquinone (HQ) exerted immunosuppressive and anti-inflammatory activity, various HQ derivatives have been synthesized and their biological activities investigated. In this study, we explored the anti-inflammatory activity of JS-III-49, a novel HQ derivative, in macrophage-mediated inflammatory responses. JS-III-49 suppressed the production of the inflammatory mediators nitric oxide (NO) and prostaglandin $E_2$ ($PGE_2$) and down-regulated the mRNA expression of the inflammatory enzymes cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) as well as the expression of the pro-inflammatory cytokines interleukin-6 (IL-6) and IL-$1{\beta}$ without cytotoxicity in LPS-stimulated RAW264.7 cells. JS-III-49 inhibited nuclear translocation of the $NF-{\kappa}B$ transcription factors p65 and p50 by directly targeting Akt, an upstream kinase of the $NF-{\kappa}B$ pathway, in LPS-stimulated RAW264.7 cells. However, JS-III-49 did not directly inhibit the kinase activities of Src and Syk, which are upstream kinases of Akt, in LPS-stimulated RAW264.7 cells. Moreover, JS-III-49 suppressed the nuclear translocation of c-Fos, one of the components of AP-1, by specifically targeting p38, an upstream mitogen-activated protein kinase (MAPK) in the AP-1 pathway in LPS-stimulated RAW264.7 cells. These results suggest that JS-III-49 plays an anti-inflammatory role in LPS-stimulated macrophages by targeting Akt and p38 in the $NF-{\kappa}B$ and AP-1 pathways, respectively.