首页> 外文期刊>Molecular pharmacology. >β-arrestin1 and distinct CXCR4 structures are required for stromal derived factor-1 to downregulate CXCR4 cell-surface levels in neuroblastoma
【24h】

β-arrestin1 and distinct CXCR4 structures are required for stromal derived factor-1 to downregulate CXCR4 cell-surface levels in neuroblastoma

机译:β-arrestin1和独特的CXCR4结构是基质衍生因子-1下调神经母细胞瘤中CXCR4细胞表面水平所必需的

获取原文
获取原文并翻译 | 示例
       

摘要

CXC chemokine receptor 4 (CXCR4) is a G protein-coupled receptor (GPCR) located on the cell surface that signals upon binding the chemokine stromal derived factor-1 (SDF-1; also called CXCL 12). CXCR4 promotes neuroblastoma proliferation and chemotaxis. CXCR4 expression negatively correlates with prognosis and drives neuroblastoma growth and metastasis in mouse models. All functions of CXCR4 require its expression on the cell surface, yet the molecular mechanisms that regulate CXCR4 cell-surface levels in neuroblastoma are poorly understood. We characterized CXCR4 cell-surface regulation in the related SH-SY5Y and SK-N-SH human neuroblastoma cell lines. SDF-1 treatment caused rapid down-modulation of CXCR4 in SH-SY5Y cells. Pharmacologic activation of protein kinase C similarly reduced CXCR4, but via a distinct mechanism. Analysis of CXCR4 mutants delineated two CXCR4 regions required for SDF-1 treatment to decrease cell-surface CXCR4 in neuroblastoma cells: the isoleucine-leucine motif at residues 328 and 329 and residues 343-352. In contrast, and unlike CXCR4 regulation in other cell types, serines 324, 325, 338, and 339 were not required. Arrestin proteins can bind and regulate GPCR cell-surface expression, often functioning together with kinases such as G protein-coupled receptor kinase 2 (GRK2). Using SK-N-SH cells which are naturally deficient in β-arrestin1, we showed that β-arrestin1 is required for the CXCR4 343-352 region to modulate CXCR4 cell-surface expression following treatment with SDF-1. Moreover, GRK2 overexpression enhanced CXCR4 internalization, via a mechanism requiring both β-arrestin1 expression and the 343-352 region. Together, these results characterize CXCR4 structural domains and β-arrestin1 as critical regulators of CXCR4 cell-surface expression in neuroblastoma. β-Arrestin1 levels may therefore influence the CXCR4-driven metastasis of neuroblastoma as well as prognosis.
机译:CXC趋化因子受体4(CXCR4)是位于细胞表面的G蛋白偶联受体(GPCR),在结合趋化因子基质衍生因子1(SDF-1;也称为CXCL 12)后发出信号。 CXCR4促进神经母细胞瘤增殖和趋化性。在小鼠模型中,CXCR4表达与预后负相关,并驱动神经母细胞瘤生长和转移。 CXCR4的所有功能都需要在细胞表面表达,但对神经母细胞瘤中调节CXCR4细胞表面水平的分子机制了解甚少。我们在相关的SH-SY5Y和SK-N-SH人神经母细胞瘤细胞系中表征了CXCR4细胞表面调节。 SDF-1处理导致SH-SY5Y细胞中CXCR4迅速下调。蛋白激酶C的药理活化类似地降低了CXCR4,但通过不同的机制。 CXCR4突变体的分析描述了SDF-1处理减少神经母细胞瘤细胞中细胞表面CXCR4所需的两个CXCR4区域:残基328和329和残基343-352的异亮氨酸-亮氨酸基序。相反,与其他细胞类型中的CXCR4调节不同,不需要丝氨酸324、325、338和339。抑制蛋白可以结合并调节GPCR细胞表面的表达,通常与诸如G蛋白偶联受体激酶2(GRK2)之类的激酶一起发挥作用。使用自然缺乏β-arrestin1的SK-N-SH细胞,我们显示,在用SDF-1处理后,CXCR4 343-352区域需要β-arrestin1来调节CXCR4细胞表面表达。此外,GRK2过表达通过同时需要β-arrestin1表达和343-352区的机制增强了CXCR4的内在化。总之,这些结果将CXCR4结构域和β-arrestin1表征为神经母细胞瘤CXCR4细胞表面表达的关键调节剂。因此,β-Arrestin1水平可能会影响CXCR4驱动的神经母细胞瘤转移以及预后。

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