首页> 外文期刊>PLoS Genetics >mTOR acts as a pivotal signaling hub for neural crest cells during craniofacial development
【24h】

mTOR acts as a pivotal signaling hub for neural crest cells during craniofacial development

机译:MTOR在颅面发育期间用作神经嵴细胞的枢转信号毂

获取原文
       

摘要

mTOR is a highly conserved serine/threonine protein kinase that is critical for diverse cellular processes in both developmental and physiological settings. mTOR interacts with a set of molecules including Raptor and Rictor to form two distinct functional complexes, namely the mTORC1 and mTORC2. Here, we used novel genetic models to investigate functions of the mTOR pathway for cranial neural crest cells (NCCs), which are a temporary type of cells arising from the ectoderm layer and migrate to the pharyngeal arches participating craniofacial development. mTOR deletion elicited a proliferation deficit and excessive apoptosis of post-migratory NCCs, leading to growth arrest of the facial primordia along with midline orofacial clefts. Furthermore, NCC differentiation was impaired. Thus, NCC derivatives, such as skeletons, vasculatures and neural tissues were either rudimentary or malformed. We further demonstrate that disruption of mTOR caused P53 hyperactivity and cell cycle arrest in cranial NCCs, and lowering P53 activity by one copy reduction attenuated the severity of craniofacial phenotype in NCC- mTOR knockout mice. Remarkably, NCC- Rptor disruption caused a spectrum of defects mirroring that of the NCC- mTOR deletion, whereas NCC- Rictor disruption only caused a mild craniofacial phenotype compared to the mTOR and Rptor conditional knockout models. Altogether, our data demonstrate that mTOR functions mediated by mTORC1 are indispensable for multiple processes of NCC development including proliferation, survival, and differentiation during craniofacial morphogenesis and organogenesis, and P53 hyperactivity in part accounts for the defective craniofacial development in NCC- mTOR knockout mice. Author summary mTOR is a highly conserved serine/threonine protein kinase that plays critical roles in diverse processes. mTOR acts through forming two distinct functional complexes, namely the mTORC1 and mTORC2. In this study, we generated novel genetic models to investigate the functions of the mTOR pathway for neural crest cell (NCC), which is an important embryonic cell type for craniofacial development. Disruption of mTOR elicited a proliferation deficit and excessive cell death of post-migratory NCCs, leading to facial growth deficiency and orofacial clefts. Furthermore, NCC differentiation was impaired and NCC derivatives, such as skeletons, vasculatures and neural tissues were either rudimentary or malformed. We also found that disruption of mTOR caused P53 hyperactivity, and lowering P53 activity ameliorated the craniofacial phenotype seen in NCC- mTOR knockout mice, suggesting that P53 hyperactivity might in part account for the craniofacial defects in mutant mice. Remarkably, disruption of mTORC1 by deletion of the gene encoding the core component Raptor caused a spectrum of defects mirroring that of the NCC- mTOR knockout mice, suggesting that mTOR regulates NCC development principally through the mTORC1 pathway during embryogenesis.
机译:MTOR是一种高度保守的丝氨酸/苏氨酸蛋白激酶,对于发育和生理环境中的不同细胞过程至关重要。 MTOR与一组分子相互作用,包括猛禽和RICTOR,形成两个不同的官能复合物,即MTORC1和MTORC2。在这里,我们使用了新的遗传模型来研究颅神经嵴细胞(NCCs)的MTOR途径的功能,这是一种临时类型的临时类型的细胞,并迁移到参与颅面发育的咽部曲线。 MTOR删除引发了迁移后NCC的增殖缺陷和过量凋亡,导致面部基金的生长逮捕以及中线口腔裂缝。此外,NCC分化受损。因此,诸如骨骼,血管和神经组织的NCC衍生物是基本的或畸形的。我们进一步证明,MTOR的破坏引起了颅骨NCCs中的P53多动和细胞周期停滞,并通过一个拷贝减少降低P53活性,减弱了NCC-MTOR敲除小鼠中的颅面表型的严重程度。值得注意的是,NCC-RPTOR破坏引起了镜像NCC-MTOR缺失的缺点,而NCC-Rictor破坏仅与MTOR和RPTOR条件敲除模型相比,仅引起轻度颅面表型。完全,我们的数据表明,MTORC1介导的MTOR函数对于NCC开发的多种过程是必不可少的,包括在颅面形态发生和器官发生期间的增殖,存活和分化,以及P53多动症部分占NCC-MTOR敲除小鼠的缺陷颅面发育。作者摘要MTOR是一个高度保守的丝氨酸/苏氨酸蛋白激酶,其在不同的过程中起重要作用。 MTOR通过形成两个不同的官能复合物,即MTORC1和MTORC2。在本研究中,我们产生了新的遗传模型,以研究神经嵴细胞(NCC)的MTOR途径的功能,这是用于颅面发育的重要胚胎细胞类型。 MTOR的破坏引发了迁移后NCC的增殖缺陷和过量的细胞死亡,导致面部生长缺乏和orofacial的CLEFTS。此外,NCC分化受损,并且NCC衍生物(例如骨骼,血管和神经组织)是基本的或畸形的。我们还发现,MTOR的破坏引起P53多动,降低P53活性改善了NCC-MTOR敲除小鼠中所见的颅面表型,表明P53多动可能在部分占突变小鼠的颅面缺陷的部分。值得注意的是,通过缺失编码核心组分猛杆的基因的MTORC1破坏引起了镜像NCC-MTOR敲除小鼠的缺点,表明MTOR主要通过胚胎发生期间主要通过MTORC1途径调节NCC开发。

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