首页> 外文期刊>Proceedings of the National Academy of Sciences of the United States of America >Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein
【24h】

Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein

机译:通过募集TSG101蛋白在质膜上形成和释放包含抑制蛋白域的蛋白1介导的微囊泡(ARMM)

获取原文
获取原文并翻译 | 示例
           

摘要

Mammalian cells are capable of delivering multiple types of membrane capsules extracellularly. The limiting membrane of late endosomes can fuse with the plasma membrane, leading to the extracellular release of multivesicular bodies (MVBs), initially contained within the endosomes, as exosomes. Budding viruses exploit the TSG101 protein and endosomal sorting complex required for transport (ESCRT) machinery used for MVB formation to mediate the egress of viral particles from host cells. Here we report the discovery of a virus-independent cellular process that generates microvesicles that are distinct from exosomes and which, like budding viruses, are produced by direct plasma membrane budding. Such budding is driven by a specific interaction of TSG101 with a tetrapeptide PSAP motif of an accessory protein, arrestin domain-containing protein 1 (ARRDC1), which we show is localized to the plasma membrane through its arrestin domain. This interaction results in relocation of TSG101 from endosomes to the plasma membrane and mediates the release of microvesicles that contain TSG101, ARRDC1, and other cellular proteins. Unlike exosomes, which are derived from MVBs, ARRDC1-mediated microvesicles (ARMMs) lack known late endosomal markers. ARMMs formation requires VPS4 ATPase and is enhanced by the E3 ligase WWP2, which interacts with and ubiquitinates ARRDC1. ARRDC1 protein discharged into ARMMs was observed in co-cultured cells, suggesting a role for ARMMs in intercellular communication. Our findings reveal an intrinsic cellular mechanism that results in direct budding of microvesicles from the plasma membrane, providing a formal paradigm for the evolutionary recruitment of ESCRT proteins in the release of budding viruses.
机译:哺乳动物细胞能够在细胞外递送多种类型的膜囊。晚期内体的限制膜可以与质膜融合,导致最初作为内体包含在内体中的多囊泡体(MVB)在细胞外释放。萌芽病毒利用TSG101蛋白和运输所需的内体分选复合物(ESCRT),用于MVB形成,以介导病毒颗粒从宿主细胞中流出。在这里,我们报告发现了一种与病毒无关的细胞过程,该过程产生了与囊泡不同的微泡,并且与萌芽病毒一样,是通过直接质膜萌芽产生的。这种出芽是由TSG101与辅助蛋白(包含抑制蛋白结构域蛋白1(ARRDC1))的四肽PSAP母题的特异性相互作用驱动的,我们证明该蛋白通过其抑制蛋白结构域定位于质膜。这种相互作用导致TSG101从内体重新定位到质膜,并介导包含TSG101,ARRDC1和其他细胞蛋白的微囊泡的释放。与来源于MVB的外来体不同,ARRDC1介导的微泡(ARMM)缺乏已知的晚期内体标记。 ARMM的形成需要VPS4 ATPase,并被E3连接酶WWP2增强,该酶与ARRDC1相互作用并使其泛素化。在共培养的细胞中观察到释放到ARMMs中的ARRDC1蛋白,表明ARMMs在细胞间通讯中的作用。我们的发现揭示了一种内在的细胞机制,可导致微囊泡从质膜中直接出芽,从而为发芽病毒释放中的ESCRT蛋白的进化募集提供了正式的范例。

著录项

  • 来源
  • 作者单位

    Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health;

    Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health;

    Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health;

    Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305;

    Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health Department of Genetics and Complex Diseases,Harvard School of Public Health, Boston, MA 02115;

  • 收录信息 美国《科学引文索引》(SCI);美国《生物学医学文摘》(MEDLINE);美国《化学文摘》(CA);
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类
  • 关键词

    gag; receptor; ubiquitin; vesicle;

    机译:插科打诨;受体泛素囊泡;

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