...
首页> 外文期刊>Oncogene >5q|[ndash]| myelodysplastic syndromes: chromosome 5q genes direct a tumor-suppression network sensing actin dynamics
【24h】

5q|[ndash]| myelodysplastic syndromes: chromosome 5q genes direct a tumor-suppression network sensing actin dynamics

机译:5q | [ndash] |骨髓增生异常综合症:染色体5q基因指导一个感知肌动蛋白动力学的肿瘤抑制网络

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Complete loss or interstitial deletions of chromosome 5 are the most common karyotypic abnormality in myelodysplastic syndromes (MDSs). Isolated del(5q)/5q– MDS patients have a more favorable prognosis than those with additional karyotypic defects, who tend to develop myeloproliferative neoplasms (MPNs) and acute myeloid leukemia. The frequency of unbalanced chromosome 5 deletions has led to the idea that 5q harbors one or more tumor-suppressor genes that have fundamental roles in the growth control of hematopoietic stem/progenitor cells (HSCs/HPCs). Cytogenetic mapping of commonly deleted regions (CDRs) centered on 5q31 and 5q32 identified candidate tumor-suppressor genes, including the ribosomal subunit RPS14, the transcription factor Egr1/Krox20 and the cytoskeletal remodeling protein, α-catenin. Although each acts as a tumor suppressor, alone or in combination, no molecular mechanism accounts for how defects in individual 5q candidates may act as a lesion driving MDS or contributing to malignant progression in MPN. One candidate gene that resides between the conventional del(5q)/5q– MDS-associated CDRs is DIAPH1 (5q31.3). DIAPH1 encodes the mammalian Diaphanous-related formin, mDia1. mDia1 has critical roles in actin remodeling in cell division and in response to adhesive and migratory stimuli. This review examines evidence, with a focus on mouse gene-targeting experiments, that mDia1 acts as a node in a tumor-suppressor network that involves multiple 5q gene products. The network has the potential to sense dynamic changes in actin assembly. At the root of the network is a transcriptional response mechanism mediated by the MADS-box transcription factor, serum response factor (SRF), its actin-binding myocardin family coactivator, MAL, and the SRF-target 5q gene, EGR1, which regulate the expression of PTEN and p53-family tumor-suppressor proteins. We hypothesize that the network provides a homeostatic mechanism balancing HPC/HSC growth control and differentiation decisions in response to microenvironment and other external stimuli.
机译:5号染色体的完全缺失或间质性缺失是骨髓增生异常综合症(MDSs)中最常见的核型异常。孤立的del(5q)/ 5q- MDS患者的预后要好于具有额外核型缺陷的患者,后者倾向于发展骨髓增生性肿瘤(MPN)和急性髓细胞性白血病。不平衡的5号染色体缺失的频率导致人们想到5q带有一个或多个肿瘤抑制基因,这些基因在造血干/祖细胞(HSC / HPC)的生长控制中具有基本作用。以5q31和5q32为中心的常见缺失区(CDR)的细胞遗传学定位确定了候选的肿瘤抑制基因,包括核糖体亚基RPS14,转录因子Egr1 / Krox20和细胞骨架重塑蛋白α-catenin。尽管每种都单独或组合用作肿瘤抑制物,但没有分子机制解释单个5q候选物中的缺陷如何充当病变驱动MDS或促进MPN的恶性进展。 DIAPH1(5q31.3)位于传统的与del(5q)/ 5q-MDS相关的CDR之间的一个候选基因。 DIAPH1编码哺乳动物与透照有关的形式mDia1。 mDia1在细胞分裂中的肌动蛋白重塑以及对黏附和迁移刺激的应答中起关键作用。这篇综述检查了针对小鼠基因靶向实验的证据,即mDia1是涉及多个5q基因产物的肿瘤抑制网络中的一个节点。该网络具有检测肌动蛋白装配中动态变化的潜力。网络的根本是由MADS-box转录因子,血清反应因子(SRF),其肌动蛋白结合型心肌素家族共激活因子MAL和SRF靶标5q基因EGR1介导的转录应答机制。和p53家族肿瘤抑制蛋白的表达我们假设该网络提供了一种稳态机制,可响应微环境和其他外部刺激来平衡HPC / HSC生长控制和差异化决策。

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