• 제목/요약/키워드: I$\kappa$B$\alpha$

검색결과 510건 처리시간 0.034초

TNF-α로 유도된 혈관내피세포의 혈관염증에 미치는 오적산(五積散)의 억제 효과 (Inhibitory Effects of Ojeoksan on TNF-α-induced Vascular Inflammation in Human Umbilical Vein Endothelial Cells)

  • 한병혁;윤정주;김혜윰;안유미;홍미현;손찬옥;나세원;이윤정;강대길;이호섭
    • 대한본초학회지
    • /
    • 제33권4호
    • /
    • pp.59-67
    • /
    • 2018
  • Objectives : Ojeoksan, originally recorded in an ancient Korean medicinal book named "Donguibogam" and has been used for the treatment of circulation disorder of blood which was called blood accumulation (血積) in Korean medicine. Therefore, this study was carried out to investigate the beneficial effect of OJS on vascular inflammation in HUVECs. Methods : We evaluated the effect of OJS on the expression of cell adhesion molecules and protective role in HUVEC stimulated by TNF-${\alpha}$ by using Western blot. Results : Pretreatment with OJS decreased the adhesion of HL-60 cells to TNF-${\alpha}$-induced HUVEC. OJS suppressed TNF-${\alpha}$-induced expression level of cell adhesion molecules such as intracellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1(VCAM-1), and endothelial cell selectin (E-selectin). Moreover, OJS significantly decreased TNF-${\alpha}$-induced production of intracellular reactive oxygen species (ROS); and inhibited the phosphorylation of $I{\kappa}B-{\alpha}$ in the cytoplasm compared to the experimental group. Pretreatment with OJS inhibited the trans-location of NF-${\kappa}B$ p65 to the nucleus. OJS also inhibited phosphorylation of MAPKs compared to the experimental group. OJS significantly increased the protein expression of Nrf2 and HO-1. Conclusions : Ojeoksan has a protective effect on vascular inflammation, and might be a potential therapeutic agent for early atherosclerosis.

급성 위염 동물 모델에서 감국(甘菊) 추출물과 아마인유(亞麻仁油) 혼합물의 위 점막 보호 효과 (Protective Effects of Chrysanthemi Indici Flos Extract and Flaxseed Oil Mixture on HCl/ethanol-induced Acute Gastric Lesion Mice)

  • 이진아;김수현;김민주;안정현;박해진;이우락;노성수
    • 대한본초학회지
    • /
    • 제33권6호
    • /
    • pp.19-28
    • /
    • 2018
  • Objectives : The objective of this study was to investigate the protective effect of Flaxseed oil and Chrysanthemi Indici Flos 50% ethanol extract in an HCl/ethanol induced acute gastritis model. Methods : ICR mice were divided into 6 groups; normal mice (Nor), gastritic mice with distilled water (Veh), gastritic mice with 10 mg/kg sucralfate (SC), gastritic mice with 16 g/㎏ Flaxseed oil (FO), gastritic mice with FO + 50 mg/kg Chrysanthemi Indici Flos (FCL), and gastritic mice with FO + 100 mg/kg Chrysanthemi Indici Flos (FCH). Then, mice were orally administered with 150 mM HCl/60% ethanol and caused acute gastritis. After 1 hr, mice were sacrificed, and blood and stomach tissue were collected. Results : Administration of FCL and FCH to mice prior to the induction of gastritis was found to reduce gastric injury. reactive oxygen species (ROS) and peroxy nitrite ($ONOO^-$) levels of stomach tissues were significantly decreased in FO, FCL, and FCH compared to Veh group. As results of stomach protein analyses, FCL and FCH effectively reduce inflammatory-related factors such as inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), and interleukin 1 beta ($IL-1{\beta}$) in gastric lesion mice. In addition, nuclear factor kappa B p65 ($NF-{\kappa}B$ p65) and phosphorylation inhibitor of nuclear factor kappa $B{\alpha}(p-I{\kappa}B{\alpha})$ were down-regulated in FCL and FCH administrated gastric lesion mice. Conclusions : These results suggest that FCL and FCH has an inhibitory effect against gastric injury. Therefore, FCL and FCH has the potential to be used as a natural therapeutic drug.

Intestinal anti-inflammatory activity of Sasa quelpaertensis leaf extract by suppressing lipopolysaccharide-stimulated inflammatory mediators in intestinal epithelial Caco-2 cells co-cultured with RAW 264.7 macrophage cells

  • Kim, Kyung-Mi;Kim, Yoo-Sun;Lim, Ji Ye;Min, Soo Jin;Ko, Hee-Chul;Kim, Se-Jae;Kim, Yuri
    • Nutrition Research and Practice
    • /
    • 제9권1호
    • /
    • pp.3-10
    • /
    • 2015
  • BACKGROUND/OBJECTIVES: Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, involves chronic inflammation of the gastrointestinal tract. Previously, Sasa quelpaertensis leaves have been shown to mediate anti-inflammation and anti-cancer effects, although it remains unclear whether Sasa leaves are able to attenuate inflammation-related intestinal diseases. Therefore, the aim of this study was to investigate the anti-inflammatory effects of Sasa quelpaertensis leaf extract (SQE) using an in vitro co-culture model of the intestinal epithelial environment. MATERIALS/METHODS: An in vitro co-culture system was established that consisted of intestinal epithelial Caco-2 cells and RAW 264.7 macrophages. Treatment with lipopolysaccharide (LPS) was used to induce inflammation. RESULTS: Treatment with SQE significantly suppressed the secretion of LPS-induced nitric oxide (NO), prostaglandin $E_2$ ($PGE_2$), IL-6, and IL-$1{\beta}$ in co-cultured RAW 264.7 macrophages. In addition, expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, and tumor necrosis factor (TNF)-${\alpha}$ were down-regulated in response to inhibition of $I{\kappa}B{\alpha}$ phosphorylation by SQE. Compared with two bioactive compounds that have previously been identified in SQE, tricin and P-coumaric acid, SQE exhibited the most effective anti-inflammatory properties. CONCLUSIONS: SQE exhibited intestinal anti-inflammatory activity by inhibiting various inflammatory mediators mediated through nuclear transcription factor kappa-B (NF-kB) activation. Thus, SQE has the potential to ameliorate inflammation-related diseases, including IBD, by limiting excessive production of pro-inflammatory mediators.

Sensitization of Cervical Carcinoma Cells to Paclitaxel by an IPP5 Active Mutant

  • Zeng, Qi-Yan;Huang, Yu;Zeng, Lin-Jie;Huang, Min;Huang, Yong-Qi;Zhu, Qi-Fang
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권19호
    • /
    • pp.8337-8343
    • /
    • 2014
  • Paclitaxel is one of the best anticancer agents that has been isolated from plants, but its major disadvantage is its dose-limiting toxicity. In this study, we obtained evidence that the active mutant IPP5 ($8-60hIPP5^m$), the latest member of the inhibitory molecules for protein phosphatase 1, sensitizes human cervix carcinoma cells HeLa more efficiently to the therapeutic effects of paclitaxel. The combination of $8-60hIPP5^m$ with paclitaxel augmented anticancer effects as compared to paclitaxel alone as evidenced by reduced DNA synthesis and increased cytotoxicity in HeLa cells. Furthermore, our results revealed that $8-60hIPP5^m$ enhances paclitaxel-induced G2/M arrest and apoptosis, and augments paclitaxel-induced activation of caspases and release of cytochrome C. Evaluation of signaling pathways indicated that this synergism was in part related to downregulation of NF-${\kappa}B$ activation and serine/threonine kinase Akt pathways. We noted that $8-60hIPP5^m$ downregulated the paclitaxel-induced NF-${\kappa}B$ activation, $I{\kappa}B{\alpha}$ degradation, PI3-K activity and phosphorylation of the serine/threonine kinase Akt, a survival signal which in many instances is regulated by NF-${\kappa}B$. Together, our observations indicate that paclitaxel in combination with $8-60hIPP5^m$ may provide a therapeutic advantage for the treatment of human cervical carcinoma.

RAW 264.7 대식세포에서 청뇌명신환(淸腦明神丸)에 의한 염증성 및 산화적 스트레스 반응 억제 효능 (Anti-inflammatory and Antioxidant Effects of Cheongnoimyungshin-hwan in RAW 264.7 Macrophages)

  • 손변우;이명화;황원덕
    • 대한한의학방제학회지
    • /
    • 제26권1호
    • /
    • pp.1-12
    • /
    • 2018
  • Objectives : Cheongnoimyungshin-hwan (CNMSH) is a Herbal compound prescription that is composed mainly of herbal medicines such as Ginseng Radix Alba, Angelicae Gigantis Radix, Dioscoreae Rhizoma, Longan Arillus and cornus cervi parvum, and for the purpose of improving memory and preventing dementia. Methods : In this study, it was investigated whether CNMSH could suppress inflammatory response and oxidative stress in the lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. As a result, CNMSH decreased expression of inducible nitric oxide (NO) synthase and cyclooxygenase-2, and also inhibited production of NO, prostaglandin E2. Results : This effect was associated with the suppression of the expression of p65, one of the nuclear factor-kappaB ($NF-{\kappa}B$) subunits, and increased expression of $I{\kappa}B-{\alpha}$, inhibit the $NF-{\kappa}B$ transcription factor. In addition, CNMSH significantly blocked intracellular reactive oxygen species accumulation in response to LPS stimulation. Furthermore, CNMSH increased expression of nuclear factor erythroid 2-related factor (Nrf)-2 activation and heme oxygenase (HO)-1. Conclusions : Therefore, it has been shown anti-inflammatory and antioxidant effects by inhibiting the expression and production of inflammatory mediators in LPS-stimulated macrophages, and is associated with ROS generation and is activated by Nrf2/HO-1 signaling pathway.

Resveratrol attenuates 4-hydroxy-2-hexenal-induced oxidative stress in mouse cortical collecting duct cells

  • Bae, Eun Hui;Joo, Soo Yeon;Ma, Seong Kwon;Lee, JongUn;Kim, Soo Wan
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제20권3호
    • /
    • pp.229-236
    • /
    • 2016
  • Resveratrol (RSV) may provide numerous protective effects against chronic inflammatory diseases. Due to local hypoxia and hypertonicity, the renal medulla is subject to extreme oxidative stress, and aldehyde products formed during lipid peroxidation, such as 4-hydroxy-2-hexenal (HHE), might be responsible for tubular injury. This study aimed at investigating the effects of RSV on renal and its signaling mechanisms. While HHE treatment resulted in decreased expression of Sirt1, AQP2, and nuclear factor erythroid 2-related factor 2 (Nrf2), mouse cortical collecting duct cells (M1) cells treated with HHE exhibited increased activation of p38 MAPK, extracellular signal regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and increased expression of NOX4, $p47^{phox}$, Kelch ECH associating protein 1 (Keap1) and COX2. HHE treatment also induced $NF-{\kappa}B$ activation by promoting $I{\kappa}B-{\alpha}$ degradation. Meanwhile, the observed increases in nuclear $NF-{\kappa}B$, NOX4, $p47^{phox}$, and COX2 expression were attenuated by treatment with Bay 117082, N-acetyl-l-cysteine (NAC), or RSV. Our findings indicate that RSV inhibits the expression of inflammatory proteins and the production of reactive oxygen species in M1 cells by inhibiting $NF-{\kappa}B$ activation.

L1 Cell Adhesion Molecule에 의한 대식세포 매개 염증반응의 억제 기전 분석 (L1 Cell Adhesion Molecule Suppresses Macrophage-mediated Inflammatory Responses)

  • 이영수
    • 약학회지
    • /
    • 제60권3호
    • /
    • pp.128-134
    • /
    • 2016
  • L1 cell adhesion molecule (L1CAM) is a cell surface molecule to initiate a variety of cellular responses through interacting with other cell adhesion molecules in a homophilic or heterophilic manner. Although its expression was found to be upregulated in some tumor cells, including cholangiocarcinomas, and ovarian cancers, and many studies have investigated the role of L1CAM in these cancers, its role in inflammatory responses has been poorly understood. In this study, we explored the role of L1CAM in macrophage-mediated inflammatory responses. L1CAM significantly suppressed the production of nitric oxide (NO), but induced cell proliferation in RAW264.7 cells. L1CAM expression was detectable, but its expression was markedly decreased by lipopolysaccharide (LPS) in RAW264.7 cells. In addition, the expression of pro-inflammatory genes, such as tumor necrosis factor (TNF)-${\alpha}$, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) induced by LPS was dramatically suppressed by L1CAM in RAW264.7 cells. L1CAM inhibited the transcriptional activities of NF-${\kappa}B$ and AP-1 while its cytoplasmic domain deletion form, $L1{\Delta}CD$ did not suppressed their activities in RAW264.7 cells. Moreover, L1CAM suppressed nuclear translocation of p65 and p50 as well as c-Jun, c-Fos and p-ATF2 which are transcription factors of NF-${\kappa}B$ and AP-1, respectively. In conclusion, L1CAM suppressed inflammatory responses in macrophages through inhibiting NF-${\kappa}B$ and AP-1 pathways.

잔가시 물 추출물의 항염증 효과 (Anti-inflammatory Activity of Sargassum micracanthum Water Extract)

  • 정다현;강보경;김꽃봉우리;김민지;안동현
    • Journal of Applied Biological Chemistry
    • /
    • 제57권3호
    • /
    • pp.227-234
    • /
    • 2014
  • 본 연구에서는 LPS로 유도된 RAW 264.7 대식세포의 염증반응을 통해 잔가시 모자반 (S. micracanthum) 물 추출물의 항염증 활성을 알아보았다. 잔가시 모자반 물 추출물이 대식세포에 미치는 독성을 알아보기 위해 MTT assay를 시행했으며, NO 생성량을 비롯하여 TNF-${\alpha}$, IL-6 및 IL-$1{\beta}$와 같은 염증 매개성 사이토카인 분비량을 측정하기 위해 ELISA법을 사용하였다. 또한, immunoblotting을 통해 iNOS, COX-2 및 NF-${\kappa}B$ p65 단백질 발현량을 알아보았다. 실험결과, 잔가시 모자반 물 추출물이 대식세포에 미치는 독성은 보이지 않았으며, NO 및 염증 매개성 사이토카인의 분비량이 농도 의존적으로 억제됨을 보였다. 특히, IL-$1{\beta}$ 분비량이 $50{\mu}g/mL$에서 50% 이상 저해됨을 보였다. 또한, iNOS, COX-2 및 NF-${\kappa}B$ p65 단백질 발현량에서도 농도의존적인 감소를 보였다. 모자반 물 추출물을 5,000 mg/kg body weight까지 경구투여 한 후 2주 동안 관찰한 결과 경구독성을 보이지 않음을 확인하였다. 그 후, 동물실험을 통해 염증으로 인한 귀 부종의 두께를 측정한 결과, 가장 높은 처리 농도인 250 mg/kg body weight으로 경구투여 하였을 때 prednisolone을 투여한 대조군과 유의적으로 비슷한 수준까지 귀 부종이 완화됨을 보였다. 따라서 본 연구는 잔가시 모자반 물 추출물이 뛰어난 항염증 효과를 가지고 있음을 나타내며, 향후 염증질환 치료제 개발에 잔가시 모자반 물 추출물이 이용될 수 있을 것으로 사료된다.

Inhibition of Inducible Nitric Oxide Synthase Expression by YS 49, a Synthetic Isoquinoline Alkaloid, in ROS 17/2.8 Cells Activated with $TNF-{\alpha},\;IFN-{\gamma}$ and LPS

  • Kang, Young-Jin;Kang, Sun-Young;Lee, Young-Soo;Park, Min-Kyu;Kim, Hye-Jung;Seo, Han-Geuk;Lee, Jae-Heun;YunChoi, Hye-Sook;Chang, Ki-Churl
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제8권5호
    • /
    • pp.273-280
    • /
    • 2004
  • Nitric oxide (NO) has been suggested to act as a mediator of cytokine-induced effects of turn over of bone. Activation of the inducible nitric oxide synthase (iNOS) by inflammation has been related with apoptotic cell death in osteoblast. YS 49, a synthetic isoquinoline alkaloid, inhibits NO production in macrophages activated with cytokines. In the present study, we investigated the molecular mechanism of YS 49 to inhibit iNOS expression in ROS 17/2.8 cells, which were activated with combined treatment of inflammatory cytokines $(TNF-{\alpha},\;IFN-{\gamma})$ and lipopolysaccharide (LPS). Results indicated that YS 49 concentration-dependently reduced iNOS mRNA and protein expression, as evidenced by Northern and Western blot analysis, respectively. The underlying mechanism by which YS 49 suppressed iNOS expression was not to affect iNOS mRNA stability but to inhibit activation and translocation of $NF-_kB$ by preventing the degradation of its inhibitory protein $I_kB_{\alpha}$. As expected, YS 49 prevented NO-induced apoptotic cell death by sodium nitroprusside. Taken together, it is concluded that YS 49 inhibits iNOS expression by interfering with degradation of phosphorylated inhibitory $_kB_{\alpha}\;(p-I_kB_{\alpha})$. These actions may be beneficial for the treatment of inflammation of the joint, such as rheumatoid arthritis.

In vitro antioxidative and anti-inflammatory effects of the compound K-rich fraction BIOGF1K, prepared from Panax ginseng

  • Hossen, Muhammad Jahangir;Hong, Yong Deog;Baek, Kwang-Soo;Yoo, Sulgi;Hong, Yo Han;Kim, Ji Hye;Lee, Jeong-Oog;Kim, Donghyun;Park, Junseong;Cho, Jae Youl
    • Journal of Ginseng Research
    • /
    • 제41권1호
    • /
    • pp.43-51
    • /
    • 2017
  • Background: BIOGF1K, a compound K-rich fraction prepared from the root of Panax ginseng, is widely used for cosmetic purposes in Korea. We investigated the functional mechanisms of the anti-inflammatory and antioxidative activities of BIOGF1K by discovering target enzymes through various molecular studies. Methods: We explored the inhibitory mechanisms of BIOGF1K using lipopolysaccharide-mediated inflammatory responses, reporter gene assays involving overexpression of toll-like receptor adaptor molecules, and immunoblotting analysis. We used the 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay to measure the antioxidative activity. We cotransfected adaptor molecules, including the myeloid differentiation primary response gene 88 (MyD88) and Toll/interleukin-receptor domain containing adaptor molecule-inducing interferon-${\beta}$ (TRIF), to measure the activation of nuclear factor (NF)-${\kappa}B$ and interferon regulatory factor 3 (IRF3). Results: BIOGF1K suppressed lipopolysaccharide-triggered NO release in macrophages as well as DPPH-induced electron-donating activity. It also blocked lipopolysaccharide-induced mRNA levels of interferon-${\beta}$ and inducible nitric oxide synthase. Moreover, BIOGF1K diminished the translocation and activation of IRF3 and NF-${\kappa}B$ (p50 and p65). This extract inhibited the upregulation of NF-${\kappa}B$-linked luciferase activity provoked by phorbal-12-myristate-13 acetate as well as MyD88, TRIF, and inhibitor of ${\kappa}B$ ($I{\kappa}B{\alpha}$) kinase ($IKK{\beta}$), and IRF3-mediated luciferase activity induced by TRIF and TANK-binding kinase 1 (TBK1). Finally, BIOGF1K downregulated the NF-${\kappa}B$ pathway by blocking $IKK{\beta}$ and the IRF3 pathway by inhibiting TBK1, according to reporter gene assays, immunoblotting analysis, and an AKT/$IKK{\beta}$/TBK1 overexpression strategy. Conclusion: Overall, our data suggest that the suppression of $IKK{\beta}$ and TBK1, which mediate transcriptional regulation of NF-${\kappa}B$ and IRF3, respectively, may contribute to the broad-spectrum inhibitory activity of BIOGF1K.