...
首页> 外文期刊>The Journal of biological chemistry >Histone Lysine-specific Demethylase 1 (LSD1) Protein Is Involved in Sal-like Protein 4 (SALL4)-mediated Transcriptional Repression in Hematopoietic Stem Cells
【24h】

Histone Lysine-specific Demethylase 1 (LSD1) Protein Is Involved in Sal-like Protein 4 (SALL4)-mediated Transcriptional Repression in Hematopoietic Stem Cells

机译:组蛋白赖氨酸特异性去甲基酶1(LSD1)蛋白参与造血干细胞中的SAL样蛋白4(SALL4)介导的转录抑制

获取原文
   

获取外文期刊封面封底 >>

       

摘要

The stem cell protein SALL4 plays a critical role in hematopoiesis by regulating the cell fate. In primitive hematopoietic precursors, it activates or represses important genes via recruitment of various epigenetic factors such as DNA methyltransferases, and histone deacylases. Here, we demonstrate that LSD1, a histone lysine demethylase, also participates in the trans-repressive effects of SALL4. Based on luciferase assays, the amine oxidase domain of LSD1 is important in suppressing SALL4-mediated reporter transcription. In freshly isolated adult mouse bone marrows, both SALL4 and LSD1 proteins are preferentially expressed in undifferentiated progenitor cells and co-localize in the nuclei. Further sequential chromatin immunoprecipitation assay confirmed that these two factors share the same binding sites at the promoter regions of important hematopoietic regulatory genes including EBF1, GATA1, and TNF. In addition, studies from both gain- and loss-of-function models revealed that SALL4 dynamically controls the binding levels of LSD1, which is accompanied by a reversely changed histone 3 dimethylated lysine 4 at the same promoter regions. Finally, shRNA-mediated knockdown of LSD1 in hematopoietic precursor cells resulted in altered SALL4 downstream gene expression and increased cellular activity. Thus, our data revealed that histone demethylase LSD1 may negatively regulate SALL4-mediated transcription, and the dynamic regulation of SALL4-associated epigenetic factors cooperatively modulates early hematopoietic precursor proliferation.
机译:通过调节细胞命运,干细胞蛋白SALL4在血缺陷中起着关键作用。在原始的造血前体中,通过募集各种表观遗传因子,例如DNA甲基转移酶和组蛋白脱酰酶,激活或抑制重要基因。这里,我们证明LSD1,组蛋白赖氨酸脱甲基酶也参与SALL4的反式压制效果。基于荧光素酶测定,LSD1的胺氧化酶结构域在抑制SALL4介导的报告转录方面是重要的。在新鲜分离的成年小鼠骨髓中,SALL4和LSD1蛋白均优先在未分化的祖细胞中表达并在核中共定位。进一步的顺序染色质免疫沉淀测定证实,这两个因素在包括EBF1,GATA1和TNF的重要造血调节基因的启动子区域中占有相同的结合位点。此外,来自增益和函数模型的研究表明,SALL4动态地控制LSD1的结合水平,其伴随着在相同启动子区域的反向改变的组蛋白3二甲基化赖氨酸4。最后,ShRNA介导的LSD1在造血前体细胞中的敲低产生的SALL4下游基因表达和细胞活性增加。因此,我们的数据显示,组蛋白脱甲基酶LSD1可以负调节SALL4介导的转录,并且SALL4相关的表观遗传因素的动态调节协同调节早期造血前体增殖。

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