首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
2.
3.
Previous studies have demonstrated that AKT1 and AKT3 are activated by heat shock and oxidative stress via both phosphatidylinositol 3-kinase-dependent and -independent pathways. However, the activation and role of AKT2 in the stress response have not been fully elucidated. In this study, we show that AKT2 in epithelial cells is activated by UV-C irradiation, heat shock, and hyperosmolarity as well as by tumor necrosis factor alpha (TNFalpha) through a phosphatidylinositol 3-kinase-dependent pathway. The activation of AKT2 inhibits UV- and TNF alpha-induced c-Jun N-terminal kinase (JNK) and p38 activities that have been shown to be required for stress- and TNF alpha-induced programmed cell death. Moreover, AKT2 interacts with and phosphorylates I kappa B kinase alpha. The phosphorylation of I kappa B kinase alpha and activation of NF kappa B mediates AKT2 inhibition of JNK but not p38. Furthermore, phosphatidylinositol 3-kinase inhibitor or dominant negative AKT2 significantly enhances UV- and TNF alpha-induced apoptosis, whereas expression of constitutively active AKT2 inhibits programmed cell death in response to UV and TNFalpha -induced apoptosis by inhibition of stress kinases and provide the first evidence that AKT inhibits stress kinase JNK through activation of the NF kappa B pathway.  相似文献   

4.
5.
高血糖症是糖尿病并发心血管疾病的重要危险因素. 高血糖诱导产生的活性氧(ROS)能够引起糖尿病心肌病.我们的前期工作已经证实,肌肽对高糖环境下细胞凋亡具有保护作用,但其机制尚未明确.为研究肌肽对高糖诱导的大鼠心肌细胞凋亡的抑制作用及相关信号机制,以高糖诱导心肌细胞H9c2为模型,采用Brdu-ELISA法检测细胞增殖过程中DNA合成情况,流式细胞术检测细胞凋亡率,免疫印迹实验检测JNK/c-Jun、NF-κB的磷酸化水平,RT-PCR检测TNF-α的mRNA表达. 实验结果显示,肌肽能够提高高糖损伤的H9c2细胞增殖能力,降低心肌细胞凋亡,抑制高糖激活的JNK、c-Jun和NF-κB磷酸化水平及TNFα的mRNA表达. 上述结果表明,肌肽拮抗高糖诱导的心肌细胞凋亡机制与抑制p-JNK /p-c-Jun、p-NF-κB水平和TNF-α表达有关.  相似文献   

6.
The cyclooxygenase 2 (COX-2) inhibitor celecoxib (also called celebrex), approved for the treatment of colon carcinogenesis, rheumatoid arthritis, and other inflammatory diseases, has been shown to induce apoptosis and inhibit angiogenesis. Because NF-kappa B plays a major role in regulation of apoptosis, angiogenesis, carcinogenesis, and inflammation, we postulated that celecoxib modulates NF-kappa B. In the present study, we investigated the effect of this drug on the activation of NF-kappa B by a wide variety of agents. We found that celecoxib suppressed NF-kappa B activation induced by various carcinogens, including TNF, phorbol ester, okadaic acid, LPS, and IL-1 beta. Celecoxib inhibited TNF-induced I kappa B alpha kinase activation, leading to suppression of I kappa B alpha phosphorylation and degradation. Celecoxib suppressed both inducible and constitutive NF-kappa B without cell type specificity. Celecoxib also suppressed p65 phosphorylation and nuclear translocation. Akt activation, which is required for TNF-induced NF-kappa B activation, was also suppressed by this drug. Celecoxib also inhibited the TNF-induced interaction of Akt with I kappa B alpha kinase (IKK). Celecoxib abrogated the NF-kappa B-dependent reporter gene expression activated by TNF, TNF receptor, TNF receptor-associated death domain, TNF receptor-associated factor 2, NF-kappa B-inducing kinase, and IKK, but not that activated by p65. The COX-2 promoter, which is regulated by NF-kappa B, was also inhibited by celecoxib, and this inhibition correlated with suppression of TNF-induced COX-2 expression. Besides NF-kappa B, celecoxib also suppressed TNF-induced JNK, p38 MAPK, and ERK activation. Thus, overall, our results indicate that celecoxib inhibits NF-kappa B activation through inhibition of IKK and Akt activation, leading to down-regulation of synthesis of COX-2 and other genes needed for inflammation, proliferation, and carcinogenesis.  相似文献   

7.
A family of anti-apoptotic regulators known as inhibitor of apoptosis (IAP) proteins block cell death in response to diverse stimuli. In spite of the fact that cellular IAP1and 2 (c IAP1 and 2) were discovered more than 12 years ago, their physiological roles have remained obscure. Several molecular mechanisms were proposed to explain their anti-apoptotic activity, ranging from direct inhibition and ubiquitination of pro-apoptotic molecules, to the activation of pro-survival signaling. New findings present a surprising and complex twists. On the one hand, c IAP1 and c IAP2 suppress Tumor Necrosis Factor α (TNFα) stimulated cell death by preventing formation of the TNF Receptor 1 (TNFR1) pro-apoptotic signaling complex. On the other hand, they regulate pro-survival NFκB signaling pathways: in the non-canonical pathway, by ubiquitination of NFκB-inducing kinase (NIK), and in the canonical pathway, by a yet-to-be-defined mechanism. In addition, c IAPs self-regulate their protein levels through RING domain mediated auto-ubiquitination. Here, we discuss the most recent progress in our understanding of the biological roles of c-IAPs, as well as the implications of targeting c IAPs for therapeutic intervention.  相似文献   

8.
9.
The transforming Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) activates signalling on the NF-κB axis through two distinct domains in its cytoplasmic C terminus, namely, CTAR1 (amino acids [aa] 187 to 231) and CTAR2 (aa 351 to 386). The ability of CTAR1 to activate NF-κB appears to be attributable to the direct interaction of tumor necrosis factor (TNF) receptor-associated factor 2 (TRAF2), while recent work indicates that CTAR2-induced NF-κB is mediated through its association with TNF receptor-associated death domain (TRADD). LMP1 expression also results in activation of the c-Jun N-terminal kinase (JNK) (also known as stress-activated protein kinase) cascade, an effect which is mediated exclusively through CTAR2 and can be dissociated from NF-κB induction. The organization and signalling components involved in LMP1-induced JNK activation are not known. In this study we have dissected the extreme C terminus of LMP1 and have identified the last 8 aa of the protein (aa 378 to 386) as being important for JNK signalling. Using a series of fine mutants in which single amino acids between codons 379 and 386 were changed to glycine, we have found that mutations of Pro379, Glu381, Ser383, or Tyr384 diminish the ability of LMP1 CTAR2 to engage JNK signalling. Interestingly, this region was also found to be essential for CTAR2-mediated NF-κB induction and coincides with the LMP1 amino acid sequences shown to bind TRADD. Furthermore, we have found that LMP1-mediated JNK activation is synergistically augmented by low levels of TRADD expression, suggesting that this adapter protein is critical for LMP1 signalling. TRAF2 is known to associate with TRADD, and expression of a dominant-negative N-terminal deletion TRAF2 mutant was found to partially inhibit LMP1-induced JNK activation in 293 cells. In addition, the TRAF2-interacting protein A20 blocked both LMP1-induced JNK and NF-κB activation, further implicating TRAF2 in these phenomena. While expression of a kinase-inactive mutated NF-κB-inducing kinase (NIK), a mitogen-activated protein kinase kinase kinase which also associates with TRAF2, impaired LMP1 signalling on the NF-κB axis, it did not inhibit LMP1-induced JNK activation, suggesting that these two pathways may bifurcate at the level of TRAF2. These data further define a role for TRADD and TRAF2 in JNK activation and confirm that LMP1 utilizes signalling mechanisms used by the TNF receptor/CD40 family to elicit its pleiotropic activities.  相似文献   

10.
The aim of this work was to find out whether Src kinase family and c-Jun N-terminal kinase (JNK) are involved in the ROS signaling pathway that could induce mucin MUC5AC expression in cultured cells of airway epithelia (BEAS-2B). For this purpose, the impact of cigarette smoke extract (CSE) on ROS production and MUC5AC expression in BEAS-2B cells was studied. Effects of ROS scavenger dimethylthiourea (DMTU), JNK specific inhibitor SP600125, and Src specific inhibitor PP2 in the CSE-induced ROS generation and MUC5AC expression were also assessed. A dose-dependent increase of ROS production in cells exposed to different concentrations of CSE was detected. DMTU inhibited cigarette smoke-induced Src phosphorylation, suggesting the ROS involvement in activation of Src kinase. Furthermore, SP600125 reduced the expression of MUC5AC. The activation of JNK was suppressed by PP2 but not by TACE inhibitor TAPI-1 or EGFR inhibitor PD153035. These results suggest that Src kinase participate in JNK activation and MUC5AC synthesis, which is independent of the TACE/EGFR activation. We conclude that ROS-Src-JNK signal cascade plays a particular role in cigarette smoke-induced mucin MUC5AC expression in BEAS-2B cells.  相似文献   

11.
中子属于高传能线密度电离辐射,能产生比κ射线更为严重的放射损伤,肠上皮对中子辐射高度敏感,迄今未见有关中子辐射致肠上皮细胞损伤中PI3K对NF-κB信号通路调控的研究报道.本研究旨在探讨中子照射后肠上皮细胞中PI3K对NF-κB信号通路的调控及其在中子辐射致肠上皮细胞损伤中的作用.选取肠上皮细胞系-6(intestinal epithelial cell No.6,IEC-6)进行传代培养,随机分为对照组、4Gy中子照射组和4Gy中子照射+LY294002处理组,照射组和LY294002处理组细胞采用4Gy中子均匀照射,LY294002处理组细胞在照前24h给予终浓度为10κmol/L的LY294002,各组于照射后6和24h采用MTT比色法、流式细胞术和免疫印迹(Western blot)方法检测IEC-6细胞增殖活力、凋亡与坏死率以及NF-κB信号通路相关分子NF-κB(p65),IKKκ和IκBκ的表达变化.研究发现,4Gy中子照射后6和24h,IEC-6细胞增殖活力下降,凋亡和坏死率增加;应用LY294002后IEC-6细胞增殖活力较照射组明显下降,IEC-6细胞凋亡和坏死率较照射组增加.4Gy中子照射后6和24h,IEC-6细胞NF-κB(p65)和IKKκ表达升高,IκBκ表达降低;应用LY294002后NF-κB(p65)和IKKκ表达降低,IκBκ表达升高,表明4Gy中子照射可引起IEC-6细胞增殖活力下降,凋亡和坏死率增加;PI3K可激活NF-κB信号通路,对中子辐射IEC-6细胞损伤发挥保护作用.  相似文献   

12.
Beers A  Haas MJ  Wong NC  Mooradian AD 《Biochemistry》2006,45(7):2408-2413
Plasma high-density lipoprotein and apolipoprotein AI (apoAI) levels are suppressed by tumor necrosis factor alpha. To determine the molecular mechanisms responsible for the effect of TNF alpha on the apoAI promoter activity, HepG2 cells were exposed to both genetic and pharmacological modulators of TNF alpha-mediated signaling in the presence or absence of TNF alpha. Exogenous ERK1 and ERK2 expression suppressed basal apoAI promoter activity; however, only ERK2 enhanced the ability of TNF alpha to suppress apoAI promoter activity. Exogenous expression of all three MEK isoforms (MEK1, MEK2A, and MEK2E) suppressed basal apoAI promoter activity and further aggravated TNF alpha-related apoAI promoter activity inhibition. Treatment with SB202190 (p38 MAP kinase inhibitor) alone significantly increased apoAI promoter activity; however, in the presence of TNF alpha, apoAI promoter activity was suppressed to an extent similar to that in cells not treated with SB202190. ApoAI promoter activity increased in cells treated with the specific JNK inhibitor SP600125, but unlike SB202190 treatment, the level of TNF alpha-related apoAI promoter inhibition was reduced by 50%. Similarly, the level of TNF alpha-related apoAI promoter inhibition was reduced in cells transfected with JNK1 siRNA. Finally, treatment of cells with the NF-kappaB inhibitors BAY and SN-50 or overexpression of NF-kappaB subunits p50 and p65 had no effect on the ability of TNF alpha to repress apoAI promoter activity. These results suggest that TNF alpha suppresses apoAI promoter activity through both the MEK/ERK and JNK pathways but is not mediated by either p38 MAP kinase activity or NF-kappaB activation.  相似文献   

13.
14.
Previous research from our laboratory has demonstrated a novel phenomenon whereby GPCRs play a role in inhibiting cytokine-mediated c-Jun N-terminal kinase (JNK) signalling. So far this novel phenomenon seems to have been vastly overlooked, with little research in the area. Therefore, in this study we explored this further; by assessing the potential of P2YRs to mediate inhibition of cytokine-mediated JNK signalling and related functional outcomes in human endothelial cells. We utilised primary endothelial cells, and employed the use of endogenous activators of P2YRs and well characterised pharmacological inhibitors, to assess signalling parameters mediated by P2YRs, Interleukin-1β (IL-1β), TNFα and JNK. Activation of P2YRs with adenosine tri-phosphate (ATP) resulted in a time- and concentration-dependent inhibition of IL-1β-mediated phosphorylation of JNK and associated kinase activity. The effect was specific for cytokine-mediated JNK signalling, as ATP was without effect on JNK induced by other non-specific activators (e.g. sorbitol, anisomycin), nor effective against other MAPK pathways such as p38 and the canonical NFκB cascade. Pharmacological studies demonstrated a role for the P2Y11 receptor in mediating this effect, but not the P2Y1 nor the adenosine receptors (A1, A2A, A2B & A3). The novel Gαq/11 inhibitor YM254890 and a protein kinase A (PKA) inhibitor H89 both partially reversed ATP-mediated inhibition of IL-1β-stimulated JNK indicating involvement of both Gαq/11 and Gαs mediated pathways. ATP also partially reversed IL-1β-mediated induction of cyclo‑oxygenase-2 (COX-2) and E-selectin. Collectively, these studies indicate the potential for activation of purinergic receptors to protect the endothelium from inflammatory driven JNK activation and may be a new target for inflammatory disease therapy.  相似文献   

15.
16.
Tumor necrosis factor-α (TNFα), a proinflammatory cytokine, causes vascular smooth muscle cell (VSMC) proliferation and migration and promotes inflammatory vascular lesions. Nuclear factor-kappa B (NF-κB) activation by TNFα requires endosomal superoxide production by Nox1. In endothelial cells, TNFα stimulates c-Jun N-terminal kinase (JNK), which inhibits NF-κB signaling. The mechanism by which JNK negatively regulates TNFα-induced NF-κB activation has not been defined. We hypothesized that JNK modulates NF-κB activation in VSMC, and does so via a Nox1-dependent mechanism. TNFα-induced NF-κB activation was TNFR1- and endocytosis-dependent. Inhibition of endocytosis with dominant-negative dynamin (DynK44A) potentiated TNFα-induced JNK activation, but decreased ERK activation, while p38 kinase phosphorylation was not altered. DynK44A attenuated intracellular, endosomal superoxide production in wild-type (WT) VSMC, but not in NADPH oxidase 1 (Nox1) knockout (KO) cells. siRNA targeting JNK1 or JNK2 potentiated, while a JNK activator (anisomycin) inhibited, TNFα-induced NF-κB activation in WT, but not in Nox1 KO cells. TNFα-stimulated superoxide generation was enhanced by JNK1 inhibition in WT, but not in Nox1 KO VSMC. These data suggest that JNK suppresses the inflammatory response to TNFα by reducing Nox1-dependent endosomal ROS production. JNK and endosomal superoxide may represent novel targets for pharmacologic modulation of TNFα signaling and vascular inflammation.  相似文献   

17.
18.
19.
《The Journal of cell biology》1996,133(5):1083-1093
We report here that both kappa B-dependent transactivation of a reporter gene and NF-kappa B activation in response to tumor necrosis factor (TNF alpha) or H2O2 treatments are deficient in human T47D cell transfectants that overexpress seleno-glutathione peroxidase (GSHPx). These cells feature low reactive oxygen species (ROS) levels and decreased intracellular ROS burst in response to TNF alpha treatment. Decreased ROS levels and NF-kappa B activation were likely to result from GSHPx increment since these phenomena were no longer observed when GSHPx activity was reduced by selenium depletion. The cellular contents of the two NF-kappa B subunits (p65 and p50) and of the inhibitory subunit I kappa B-alpha were unaffected by GSHPx overexpression, suggesting that increased GSHPx activity interfered with the activation, but not the synthesis or stability, of Nf-kappa B. Nuclear translocation of NF-kappa B as well as I kappa B-alpha degradation were inhabited in GSHPx-overexpressing cells exposed to oxidative stress. Moreover, in control T47D cells exposed to TNF alpha, a time correlation was observed between elevated ROS levels and I kappa B- alpha degradation. We also show that, in growing T47D cells, GSHPx overexpression altered the isoform composition of I kappa B-alpha, leading to the accumulation of the more basic isoform of this protein. GSHPx overexpression also abolished the TNF alpha-mediated transient accumulation of the acidic and highly phosphorylated I kappa B-alpha isoform. These results suggest that intracellular ROS are key elements that regulate the phosphorylation of I kappa B-alpha, a phenomenon that precedes and controls the degradation of this protein, and then NF- kappa B activation.  相似文献   

20.
TNFα is a pleotropic cytokine that initiates many downstream signaling pathways, including NF-κB activation, MAP kinase activation and the induction of both apoptosis and necrosis. TNFα has shown to lead to reactive oxygen species generation through activation of NADPH oxidase, through mitochondrial pathways, or other enzymes. As discussed, ROS play a role in potentiation or inhibition of many of these signaling pathways. We particularly discuss the role of sustained JNK activation potentiated by ROS, which generally is supportive of apoptosis and “necrotic cell death” through various mechanisms, while ROS could have inhibitory or stimulatory roles in NF-κB signaling.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号