...
首页> 外文期刊>American Journal of Physiology >Lipopolysaccharide-dependent interaction between PU.l and cJun determines production of lipocalin-type prostaglandin D synthase and prostaglandin D2 in macrophages
【24h】

Lipopolysaccharide-dependent interaction between PU.l and cJun determines production of lipocalin-type prostaglandin D synthase and prostaglandin D2 in macrophages

机译:PU.L和CJUN之间的脂多糖依赖性相互作用决定了巨噬细胞脂质素蛋白型前列腺素D合酶和前列腺素D2的产生

获取原文
获取原文并翻译 | 示例
           

摘要

Previously, we reported that expression of lipocalin-prostaglandin D synthase (L-PGDS) is inducible in macrophages and protects from Pseudomonas pneumonia. Here, we investigated the mechanism by which L-PGDS gene expression is induced in macrophages. A promoter analysis of the murine L-PGDS promoter located a binding site of PU.l, a transcription factor essential for macrophage development and inflammatory gene expression. A chromatin immunoprecipitation assay showed that PU. 1 bound to the cognate site in the endogenous L-PGDS promoter in response to LPS. Overexpression of PU.l, but not of PU.1~(S148A), a mutant inert to casein kinase II (CKII) or NF-KB-inducing kinase (NIK), induced L-PGDS in RAW 264.7 cells. Conversely, siRNA silencing of PU.l expression blunted productions of L-PGDS and prostaglandin D2 (PGD_2). LPS treatment induced formation of the complex of PU.l and cJun on the PU.l site, but inactivation of cJun by treatment with JNK or p38 kinase inhibitor abolished the complex, and suppressed PU.l transcriptional activity for L-PGDS gene expression. Together, these results show that PU.l, activated by CKII or NIK, cooperates with MAPK-activated cJun to maximally induce L-PGDS expression in macrophages following LPS treatment, and suggest that PU.l participates in innate immunity through the production of L-PGDS and PGD_2.
机译:以前,我们报道了脂素 - 前列腺素D合酶(L-PGDS)的表达在巨噬细胞中诱导,免受假单胞菌肺炎的保护。在这里,我们研究了L-PGDS基因表达在巨噬细胞中诱导的机制。鼠L-PGDS启动子的启动子分析位于PU.L的结合位点,巨噬细胞发育和炎症基因表达的转录因子。染色质免疫沉淀测定显示PU。 1响应于LPS与内源L-PGDS启动子中的同源位点结合。 PU.L的过度表达,但不具有PU.1〜(S148A),对酪蛋白激酶II(CKII)或NF-KB诱导激酶(NIK)的突变体惰性,诱导原料264.7细胞的L-PGDS。相反,SiRNA沉默于PU.L表达的沉默L-PGDS和前列腺素D2(PGD_2)的垂直。 LPS治疗诱导PU.L和Cjun的复合物的形成PU.L位点,但通过JNK或P38激酶抑制剂的治疗使Cjun失活,废除了络合物,抑制了L-PGDS基因表达的转录活性。这些结果表明,由CKII或NIK激活的PU.L与MAPK激活的CJUN合作,以最大限度地诱导LPS处理后巨噬细胞的L-PGDS表达,并建议PU.L通过生产的生产参与先天免疫力-PGDS和PGD_2。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号