...
首页> 外文期刊>Molecular and Cellular Biology >Downregulation of Protein 4.1R, a Mature Centriole Protein, Disrupts Centrosomes, Alters Cell Cycle Progression, and Perturbs Mitotic Spindles and Anaphase
【24h】

Downregulation of Protein 4.1R, a Mature Centriole Protein, Disrupts Centrosomes, Alters Cell Cycle Progression, and Perturbs Mitotic Spindles and Anaphase

机译:下调蛋白质4.1R(一种成熟的中心蛋白),打乱中心体,改变细胞周期进程,干扰有丝分裂纺锤体和后期

获取原文

摘要

Centrosomes nucleate and organize interphase microtubules and are instrumental in mitotic bipolar spindle assembly, ensuring orderly cell cycle progression with accurate chromosome segregation. We report that the multifunctional structural protein 4.1R localizes at centrosomes to distal/subdistal regions of mature centrioles in a cell cycle-dependent pattern. Significantly, 4.1R-specific depletion mediated by RNA interference perturbs subdistal appendage proteins ninein and outer dense fiber 2/cenexin at mature centrosomes and concomitantly reduces interphase microtubule anchoring and organization. 4.1R depletion causes G1 accumulation in p53-proficient cells, similar to depletion of many other proteins that compromise centrosome integrity. In p53-deficient cells, 4.1R depletion delays S phase, but aberrant ninein distribution is not dependent on the S-phase delay. In 4.1R-depleted mitotic cells, efficient centrosome separation is reduced, resulting in monopolar spindle formation. Multipolar spindles and bipolar spindles with misaligned chromatin are also induced by 4.1R depletion. Notably, all types of defective spindles have mislocalized NuMA (nuclear mitotic apparatus protein), a 4.1R binding partner essential for spindle pole focusing. These disruptions contribute to lagging chromosomes and aberrant microtubule bridges during anaphase/telophase. Our data provide functional evidence that 4.1R makes crucial contributions to the structural integrity of centrosomes and mitotic spindles which normally enable mitosis and anaphase to proceed with the coordinated precision required to avoid pathological events.
机译:中心体形成核并组织相间微管,并在有丝分裂双极纺锤体组装中发挥作用,从而确保细胞周期有序地进行,并具有精确的染色体分离。我们报道多功能细胞结构蛋白4.1R定位在中心体到成熟中心体的远端/近端区域在细胞周期依赖模式。值得注意的是,由RNA干扰介导的4.1R特异性耗竭扰乱了位于成熟中心体的近端附睾蛋白9in和外部密集纤维2 / cenexin,并同时减少了相间微管的锚定和组织。 4.1R耗竭会导致p53精巢细胞中G 1 积累,这与许多其他损害中心体完整性的蛋白质的耗竭相似。在p53缺失的细胞中,4.1R耗竭会延迟S期,但9in分布异常并不取决于S期延迟。在4.1R耗竭的有丝分裂细胞中,有效的中心体分离减少,导致单极纺锤体形成。染色质未对准的多极纺锤体和双极纺锤体也被4.1R耗尽诱导。值得注意的是,所有类型的有缺陷的纺锤体都有错误定位的NuMA( nu clear m itotic a pparatus蛋白),这是纺锤体极必不可少的4.1R结合伴侣专注。这些破坏在后期/末期导致染色体滞后和微管桥异常。我们的数据提供了功能证据,表明4.1R对中心体和有丝分裂纺锤体的结构完整性做出了重要贡献,这通常使有丝分裂和后期能够以避免病理事件所需的协调精度进行。

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