首页> 美国卫生研究院文献>Cell Death and Differentiation >Oxidative stress-induced S100B accumulation converts myoblasts into brown adipocytes via an NF-κB/YY1/miR-133 axis and NF-κB/YY1/BMP-7 axis
【2h】

Oxidative stress-induced S100B accumulation converts myoblasts into brown adipocytes via an NF-κB/YY1/miR-133 axis and NF-κB/YY1/BMP-7 axis

机译:氧化应激诱导的S100B积累通过NF-κB/ YY1 / miR-133轴和NF-κB/ YY1 / BMP-7轴将成肌细胞转化为棕色脂肪细胞

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Muscles of sarcopenic people show hypotrophic myofibers and infiltration with adipose and, at later stages, fibrotic tissue. The origin of infiltrating adipocytes resides in fibro-adipogenic precursors and nonmyogenic mesenchymal progenitor cells, and in satellite cells, the adult stem cells of skeletal muscles. Myoblasts and brown adipocytes share a common Myf5+ progenitor cell: the cell fate depends on levels of bone morphogenetic protein 7 (BMP-7), a TGF-β family member. S100B, a Ca2+-binding protein of the EF-hand type, is expressed at relatively high levels in myoblasts from sarcopenic humans and exerts anti-myogenic effects via NF-κB-dependent inhibition of MyoD, a myogenic transcription factor acting upstream of the essential myogenic factor, myogenin. Adipogenesis requires high levels of ROS, and myoblasts of sarcopenic subjects show elevated ROS levels. Here we show that: (1) ROS overproduction in myoblasts results in upregulation of S100B levels via NF-κB activation; and (2) ROS/NF-κB-induced accumulation of S100B causes myoblast transition into brown adipocytes. S100B activates an NF-κB/Ying Yang 1 axis that negatively regulates the promyogenic and anti-adipogenic miR-133 with resultant accumulation of the brown adipogenic transcription regulator, PRDM-16. S100B also upregulates BMP-7 via NF-κB/Ying Yang 1 with resultant BMP-7 autocrine activity. Interestingly, myoblasts from sarcopenic humans show features of brown adipocytes. We also show that S100B levels and NF-κB activity are elevated in brown adipocytes obtained by culturing myoblasts in adipocyte differentiation medium and that S100B knockdown or NF-κB inhibition in myoblast-derived brown adipocytes reconverts them into fusion-competent myoblasts. At last, interstitial cells and, unexpectedly, a subpopulation of myofibers in muscles of geriatric but not young mice co-express S100B and the brown adipocyte marker, uncoupling protein-1. These results suggest that S100B is an important intracellular molecular signal regulating Myf5+ progenitor cell differentiation into fusion-competent myoblasts or brown adipocytes depending on its levels.
机译:肌肉缺乏症的人的肌肉表现出营养不足的肌纤维,并伴有脂肪浸润,并在以后阶段出现纤维化组织。浸润的脂肪细胞起源于纤维脂肪形成的前体和非肌源性间充质祖细胞,而卫星细胞则是成年的骨骼肌干细胞。成肌细胞和褐色脂肪细胞共享一个共同的Myf5 + 祖细胞:细胞的命运取决于TGF-β家族成员骨形态发生蛋白7(BMP-7)的水平。 S100B是EF-hand型的Ca 2 + 结合蛋白,在肌少肌症患者的成肌细胞中以相对较高的水平表达,并通过NF-κB依赖性的MyoD抑制作用产生抗成肌作用,在必需的成肌因子肌生成素上游起作用的成肌转录因子。脂肪生成需要高水平的ROS,而少肌症患者的成肌细胞显示ROS升高。在这里,我们显示:(1)成肌细胞中ROS的过量产生通过NF-κB激活导致S100B水平上调; (2)ROS /NF-κB诱导的S100B积累导致成肌细胞转变为棕色脂肪细胞。 S100B激活一个NF-κB/ Ying Yang 1轴,该轴负调控生肌和抗脂肪生成miR-133,并导致褐色脂肪生成转录调节剂PRDM-16积累。 S100B还通过NF-κB/ Ying Yang 1上调BMP-7,从而产生BMP-7自分泌活性。有趣的是,来自肌肉减少症的人的成肌细胞显示出褐色脂肪细胞的特征。我们还表明,通过在脂肪细胞分化培养基中培养成肌细胞而获得的棕色脂肪细胞中,S100B水平和NF-κB活性升高,并且成肌细胞衍生的棕色脂肪细胞中的S100B敲低或NF-κB抑制作用将它们转化为具有融合能力的成肌细胞。最后,在老年小鼠的肌肉中,间质细胞和出乎意料的肌纤维亚群共表达S100B和棕色脂肪细胞标志物,解偶联蛋白-1。这些结果表明,S100B是调节Myf5 + 祖细胞向融合型成肌细胞或棕色脂肪细胞分化的重要细胞内分子信号。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

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