首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 203 毫秒
1.
肿瘤蛋白p53诱导的核蛋白2(TP53INP2),也称糖尿病与肥胖调控蛋白(DOR),在骨骼肌、心肌和脑等代谢旺盛的组织中表达水平较高。TP53INP2在细胞核内主要发挥转录辅激活因子的作用,如作为甲状腺激素受体的辅激活因子调控甲状腺激素相关基因的表达。后续研究发现,TP53INP2为细胞内重要的分解代谢途径—细胞自噬所必需,饥饿时TP53INP2出核参与自噬的起始。在骨骼肌中,TP53INP2通过促进自噬导致肌肉流失,而在白色脂肪组织中,TP53INP2则通过促进自噬抑制脂肪前体细胞的分化。此外,在营养丰富的条件下,TP53INP2能够定位于核仁,协助r DNA转录起始前复合物的组装,促进r DNA的转录,参与细胞内重要的合成代谢—核糖体的生物发生。临床统计数据表明,TP53INP2的表达水平与糖尿病和某些类型的癌症早期的发生发展密切相关。本文对TP53INP2在转录调控、细胞自噬和相关疾病如癌症与糖尿病中的生物学功能进行综述。  相似文献   

2.
自噬是指由膜包裹的自噬体与溶酶体融合形成自噬溶酶体,并在其中降解所包裹的内含物的过程。自噬包含自噬起始、自噬体的形成、自噬体的成熟和溶酶体的再生四个基本过程,目前已发现几十种Atg蛋白参与其中。此外,自噬受到m TOR、PI3K和ULK1的中心调控以及转录因子和micro RNA的调控。自噬对维持细胞内环境的稳定起着关键性作用,在肿瘤的形成与发展中发挥着复杂的作用。  相似文献   

3.
细胞自噬是细胞内高度保守的细胞自我消化和分解代谢过程,细胞内变性蛋白、衰老和受损的细胞器被转运到溶酶体降解. 自噬过程失调引起多种疾病,包括感染、衰老、神经退行性疾病、癌症和心脏疾病等,因此,自噬过程需要非常精确的调控. MicroRNA是一类在基因转录后水平调控目的基因的功能性小RNA分子.研究发现,microRNA可以通过RNA干扰(RNA interference, RNAi)途径调控某些自噬相关基因(autophagy related gene, ATG)及其调节因子.这些microRNA表达异常足以影响自噬水平,使得microRNA成为自噬研究的新视角,同时也使microRNA成为治疗自噬失调引起的疾病的潜在靶点.本文将对有关microRNA参与细胞自噬调控的最新研究动态进行综述.  相似文献   

4.
核受体超家族介导基因调控的分子机制   总被引:2,自引:0,他引:2  
Wang BH 《生理科学进展》2003,34(4):369-372
核受体超家族由甾体激素、甲状腺激素、维甲酸、维生素D等化学信号的受体及配体未明的多种孤儿受体组成,该家族成员的主要功能是作为配体激活的转录因子,调控代谢、发育、生殖相关基因的表达。核受体与启动子和增强子上的激素应答元件及其它DNA序列特异性激活因子结合,而激活或阻遏靶基因的转录。核受体调控基因转录需要募集称为辅调控因子的蛋白分子,这些蛋白分子与核受体一起装配成多组分的复合物,它们可提供相关的酶促活性和脚手架功能。通过与基础转录机器的相互作用和对染色质结构的可逆性共价修饰等作用,辅调控因子调控核受体对靶基因转录的激活或阻遏。许多辅调控因子本身受到多条细胞内信号转导途径的调控。  相似文献   

5.
TP53基因(编码p53蛋白)作为一个重要的抑瘤基因,通过调控一系列信号转导通路广泛参与了多种恶性肿瘤的发生发展,一直是肿瘤分子生物学研究领域的热点.最近的研究发现,microRNAs(miRNAs)参与了TP53的信号通路,它们之间存在着复杂的调控网络.一方面,p53通过调控一些miRNAs的转录及转录后成熟,促进细胞周期阻滞、诱导细胞凋亡和衰老,抑制肿瘤发生.另一方面,许多miRNAs,如miR-25、miR-30d、miR-125b和miR-504等可直接调控p53的表达与活性,参与TP53信号通路的调节,还有一些miRNAs则通过调节p53上下游基因,发挥重要的生物学功能.其中,最具有代表性的是miR-34家族,它们受p53直接调控并参与TP53信号通路,通过靶向抑制多个TP53信号通路关键分子的表达,发挥抑瘤作用.此外,它们还可以通过抑制沉默信息调节子,增强p53的活性,反馈调节TP53信号通路.miRNAs与TP53之间调控网络的研究,是对TP53抑瘤机制的重要补充.  相似文献   

6.
为深入研究肿瘤蛋白p53诱导核蛋白1(Tumor protein 53-induced nuclear protein 1, TP53INP1)的结构及其在微囊藻毒素-LR(MC-LR)胁迫下的表达变化,以MC-LR诱导的草鱼(Ctenopharygodon idella)肝脏转录组测序获得的unigenes序列为基础,扩增获得了TP53INP1基因的cDNA序列(GenBank登录号:MG797689),其中开放阅读框(Open reading frame, ORF)为759 bp,编码252个氨基酸,属于β类型,具有4个PEST结构。氨基酸同源性分析结果表明, TP53INP1具有较高的保守性,其中与鱇浪白鱼(Anabarilius grahami)相似性最高。系统进化分析结果表明其与鱇浪白鱼(Anabarilius grahami)、斑马鱼(Danio rerio)等鱼类聚为一大支。采用荧光定量PCR分析发现TP53INP1基因在草鱼各组织中广泛分布,其中,在肝脏和血液等组织中表达丰富,显著高于在头肾组织中的表达(P<0.05)。采用Western blot检测分析不同剂量...  相似文献   

7.
肿瘤抑制因子p53主要作为转录因子发挥作用.当细胞受到诸如缺氧、DNA损伤等胁迫时,p53蛋白迅速在细胞内积聚并激活,从而调控一系列基因的转录,导致细胞周期停顿、凋亡或衰老,避免细胞癌化.p53功能的失活往往导致癌症发生.编码p53蛋白的TP53基因的突变是p53失活的主要方式.突变型p53不仅失去抑癌作用,而且还具有...  相似文献   

8.
骨骼肌由异质性的肌纤维组成,不同类型的肌纤维具有不同的形态、代谢、生理和生化特性.根据不同肌纤维中表达的特异肌球蛋白重链亚型可将成体哺乳动物骨骼肌纤维分为4类,即Ⅰ,Ⅱa,Ⅱx和Ⅱb型.骨骼肌保持高度可塑性,当机体受到某些生理或病理刺激时,骨骼肌为了适应需要,通过激活胞内相关信号通路改变肌纤维特异基因的表达从而诱发肌纤维类型的转化.本文综述了细胞内参与调控肌纤维类型转化的多条重要信号通路,如Ca2+信号通路,Ras/MAPK信号通路及多种转录调节因子,辅激活因子和抑制子等,为改善肉类品质,提高运动训练效果及治疗肌肉相关疾病奠定了理论基础.  相似文献   

9.
细胞自噬是真核细胞中广泛存在的一种自我保护机制,是细胞在应激情况下通过溶酶体或液泡高度保守的降解途径将细胞内异常蛋白和细胞器降解为生物大分子,重新被细胞利用的过程。适度的运动锻炼可以诱导机体多种组织细胞自噬的激活,增强机体的活力,延缓机体的衰老。运动训练可以刺激骨骼肌细胞自噬水平上调,延缓骨骼肌衰老;运动训练作为一种机械性刺激可以通过调节心肌细胞的自噬激活调控长寿命或错误折叠心肌蛋白和受损细胞器的代谢,延缓心肌衰老;此外,细胞自噬与糖尿病、肿瘤、脑血管疾病、衰老及心脏病等密切相关,运动训练可以预防动脉粥样硬化等血管类疾病的发生,也可以通过调控细胞自噬来预防与治疗心脏病、中风、糖尿病等疾病。现主要论述细胞自噬的涵义与分类,细胞自噬不同阶段的分子机制,以及运动训练通过调控细胞自噬相关基因调控骨骼肌、心肌和自噬相关疾病的分子机制,为使用科学的运动训练方式来提高机体功能及预防和治疗疾病提供了理论依据。  相似文献   

10.
细胞自噬是一种进化上保守的分解代谢过程,涉及细胞内长寿命蛋白和受损伤细胞器的降解,其在细胞内稳态、肿瘤、心力衰竭、衰老相关性疾病、神经退行性疾病以及传染病等多种生命进程中发挥着重要作用。泛素样蛋白系统、m TOR信号通路、micro RNA、caspase等均参与了细胞自噬调控过程。该文综述了细胞自噬过程、功能和分子调控机制的研究进展,以期有助于研究细胞自噬机理,为治疗心脏疾病(如动脉粥样硬化)、癌症(如乳腺癌)等提供理论基础。  相似文献   

11.
Using a bioinformatic approach, we identified a TP53INP1-related gene encoding a protein with 30% identity with tumor protein 53-induced nuclear protein 1 (TP53INP1), which was named TP53INP2. TP53INP1 and TP53INP2 sequences were found in several species ranging from Homo sapiens to Drosophila melanogaster, but orthologues were found neither in earlier eukaryotes nor in prokaryotes. To gain insight into the function of the TP53INP2 protein, we carried out a yeast two-hybrid screening that showed that TP53INP2 binds to the LC3-related proteins GABARAP and GABARAP-like2, and then we demonstrated by coimmunoprecipitation that TP53INP2 interacts with these proteins, as well as with LC3 and with the autophagosome transmembrane protein VMP1. TP53INP2 translocates from the nucleus to the autophagosome structures after activation of autophagy by rapamycin or starvation. Also, we showed that TP53INP2 expression is necessary for autophagosome development because its small interfering RNA-mediated knockdown strongly decreases sensitivity of mammalian cells to autophagy. Finally, we found that interactions between TP53INP2 and LC3 or the LC3-related proteins GABARAP and GABARAP-like2 require autophagy and are modulated by wortmannin as judged by bioluminescence resonance energy transfer assays. We suggest that TP53INP2 is a scaffold protein that recruits LC3 and/or LC3-related proteins to the autophagosome membrane by interacting with the transmembrane protein VMP1. It is concluded that TP53INP2 is a novel gene involved in the autophagy of mammalian cells.  相似文献   

12.
13.
TP53INP1 (tumor protein 53-induced nuclear protein 1) is a tumor suppressor, whose expression is downregulated in cancers from different organs. It was described as a p53 target gene involved in cell death, cell-cycle arrest and cellular migration. In this work, we show that TP53INP1 is also able to interact with ATG8-family proteins and to induce autophagy-dependent cell death. In agreement with this finding, we observe that TP53INP1, which is mainly nuclear, relocalizes in autophagosomes during autophagy where it is eventually degraded. TP53INP1-LC3 interaction occurs via a functional LC3-interacting region (LIR). Inactivating mutations of this sequence abolish TP53INP1-LC3 interaction, relocalize TP53INP1 in autophagosomes and decrease TP53INP1 ability to trigger cell death. Interestingly, TP53INP1 binds to ATG8-family proteins with higher affinity than p62, suggesting that it could partially displace p62 from autophagosomes, modifying thereby their composition. Moreover, silencing the expression of autophagy related genes (ATG5 or Beclin-1) or inhibiting caspase activity significantly decreases cell death induced by TP53INP1. These data indicate that cell death observed after TP53INP1-LC3 interaction depends on both autophagy and caspase activity. We conclude that TP53INP1 could act as a tumor suppressor by inducing cell death by caspase-dependent autophagy.  相似文献   

14.
Cells control their metabolism through modulating the anabolic and catabolic pathways. TP53INP2/DOR (tumor protein p53 inducible nuclear protein 2), participates in cell catabolism by serving as a promoter of autophagy. Here we uncover a novel function of TP53INP2 in protein synthesis, a major biosynthetic and energy-consuming anabolic process. TP53INP2 localizes to the nucleolus through its nucleolar localization signal (NoLS) located at the C-terminal domain. Chromatin immunoprecipitation (ChIP) assays detected an association of TP53INP2 with the ribosomal DNA (rDNA), when exclusion of TP53INP2 from the nucleolus repressed rDNA promoter activity and the production of ribosomal RNA (rRNA) and proteins. The removal of TP53INP2 also impaired the association of the POLR1/RNA polymerase I preinitiation complex (PIC) with rDNA. Further, TP53INP2 interacts directly with POLR1 PIC, and is required for the assembly of the complex. These data indicate that TP53INP2 promotes ribosome biogenesis through facilitating rRNA synthesis at the nucleolus, suggesting a dual role of TP53INP2 in cell metabolism, assisting anabolism on the nucleolus, and stimulating catabolism off the nucleolus.  相似文献   

15.
16.
《Autophagy》2013,9(3):383-384
We identified a TP53INP1-related gene, which was named TP53INP2, using a bioinformatic approach. TP53INP1 and TP53INP2 sequences were found in higher eukaryotes but not in lower eukaryotes nor in prokaryotes. To gain insight into the function of the TP53INP2 protein, we carried out a number of protein-protein interaction approaches which showed that TP53INP2 binds to the Atg8-like proteins, as well as with the autophagosome trans-membrane protein VMP1. TP53INP2 translocates from the nucleus to autophagosomes after activation of autophagy. We also showed that TP53INP2 expression is necessary for autophagosome development since its siRNA-mediated knock-down strongly decreases sensitivity to induced-autophagy. We suggest that TP53INP2 is a scaffold protein that recruits Atg8-like proteins to the autophagosome membrane by interacting with the trans-membrane protein VMP1.  相似文献   

17.
18.
Tumour protein p53‐inducible nuclear protein 1 (TP53INP1) is a tumour suppressor associated with malignant tumour metastasis. Vasculogenic mimicry (VM) is a new tumour vascular supply pattern that significantly influences tumour metastasis and contributes to a poor prognosis. However, the molecular mechanism of the relationship between TP53INP1 and breast cancer VM formation is unknown. Here, we explored the underlying mechanism by which TP53INP1 regulates VM formation in vitro and in vivo. High TP53INP1 expression was not only negatively correlated with a poor prognosis but also had a negative relationship with VE‐cadherin, HIF‐1α and Snail expression. TP53INP1 overexpression inhibited breast cancer invasion, migration, epithelial‐mesenchymal transition (EMT) and VM formation; conversely, TP53INP1 down‐regulation promoted these processes in vitro by functional experiments and Western blot analysis. We established a hypoxia model induced by CoCl2 and assessed the effects of TP53INP1 on hypoxia‐induced EMT and VM formation. In addition, we confirmed that a reactive oxygen species (ROS)‐mediated signalling pathway participated in TP53INP1‐mediated VM formation. Together, our results show that TP53INP1 inhibits hypoxia‐induced EMT and VM formation via the ROS/GSK‐3β/Snail pathway in breast cancer, which offers new insights into breast cancer clinical therapy.  相似文献   

19.
Tumor protein p53-induced nuclear protein 1 (TP53INP1) is a well known stress-induced protein that plays a role in both cell cycle arrest and p53-mediated apoptosis. Loss of TP53INP1 expression has been reported in human melanoma, breast carcinoma, and gastric cancer. However, TP53INP1 expression and its regulatory mechanism in esophageal squamous cell carcinoma (ESCC) remain unclear. Our findings are in agreement with previous reports in that the expression of TP53INP1 was downregulated in 28% (10/36 cases) of ESCC lesions, and this was accompanied by significant promoter methylation. Overexpression of TP53INP1 induced G1 cell cycle arrest and increased apoptosis in ESCC cell lines (EC-1, EC-109, EC-9706). Furthermore, our study showed that the oncoprotein c-Myc bound to the core promoter of TP53INP1 and recruited DNA methyltransferase 3A to methylate the local promoter region, leading to the inhibition of TP53INP1 expression. Our findings revealed that TP53INP1 is a tumor suppressor in ESCC and that c-Myc-mediated DNA methylation-associated silencing of TP53INP1 contributed to the pathogenesis of human ESCC.  相似文献   

20.

Background

MiR-155 has emerged as an “oncomiR”, which is the most significantly up-regulated miRNA in breast cancer. However, the mechanisms of miR-155 functions as an oncomiR are mainly unknown. In this study, the aims were to investigate the effects of miR-155 on cell proliferation, cell cycle, and cell apoptosis of ERalpha (+) breast cancer cells and to verify whether TP53INP1 (tumor protein 53-induced nuclear protein 1) is a target of miR-155, and tried to explore the mechanisms of miR-155 in this process.

Results

The expression of miR-155 is significantly higher in MCF-7 cells compared with MDA-MB-231 cells. Ectopic expression of TP53INP1 inhibits growth of MCF-7 cells by inducing cell apoptosis and inhibiting cell cycle progression. Overexpression of miR-155 increases cell proliferation and suppress cell apoptosis, whereas abrogating expression of miR-155 suppress cell proliferation and promotes cell apoptosis of MCF-7 cells. In addition, miR-155 negatively regulates TP53INP1 mRNA expression and the protein expression of TP53INP1, cleaved-caspase-3, -8, -9, and p21, and luciferase reporter reveals that TP53INP1 is targeted by miR-155.

Conclusions

TP53INP1 is the direct target of miR-155. MiR-155, which is overexpressed in MCF-7 cells, contributes to proliferation of MCF-7 cells possibly through down-regulating target TP53INP1.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号